1
|
Ubeysinghe S, Sebilleau CO, Thotamune W, Rajarathna C, Azibere S, Tennakoon M, Payton JL, Sprague RS, Martin RS, Sucheck SJ, Karunarathne A. Recombinant-Chemosynthetic Biosensors for Probing Cell Surface Signaling of Red Blood Cells and Other Cells. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:95-110. [PMID: 40018647 PMCID: PMC11863169 DOI: 10.1021/cbmi.4c00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 03/01/2025]
Abstract
The complex signaling mechanisms in red blood cells (RBCs) enable them to adapt to physiological stresses such as exposure to low O2 levels, metabolic demands, oxidative stress, and shear stress. Since Ca2+ is a crucial determinant of RBC fate, various ion channels, pumps, and exchangers regulate the delicate balance of Ca2+ influx and efflux in RBCs. Elevated intracellular Ca2+ can activate processes such as membrane phospholipid scrambling and alter RBC deformability, which is essential for effective capillary transit. However, the dynamic information about Ca2+ regulation in RBCs is limited. Although static mapping and bioanalytical methods have been utilized, the absence of a nucleus and the presence of hemoglobin create challenges for real-time probing of RBC signaling, necessitating innovative approaches. This work introduces a synthetic chemistry-recombinant protein-based strategy to assemble sensors at genetically intact healthy human RBC surfaces for measuring dynamic signaling. Using this approach, we measured autocrine regulation of RBC Ca2+ influx in response to low O2 tension-induced ATP release. The study also explores the utilization of synthetic glycosylphosphatidylinositol (GPI) anchor mimics and sortagging for targeting sensors to the surfaces of primary as well as immortalized cells. This demonstrated the wide applicability of this approach to probe dynamic signaling in intact cells.
Collapse
Affiliation(s)
- Sithurandi Ubeysinghe
- Department
of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
- Institute
for Drug and Biotherapeutic Innovation, Saint Louis University, Saint
Louis, Missouri 63103, United States
| | - Chloe O. Sebilleau
- Department
of Chemistry and Biochemistry, The University
of Toledo, Toledo, Ohio 43606, United States
| | - Waruna Thotamune
- Department
of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
- Institute
for Drug and Biotherapeutic Innovation, Saint Louis University, Saint
Louis, Missouri 63103, United States
| | - Chathuri Rajarathna
- Department
of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
- Institute
for Drug and Biotherapeutic Innovation, Saint Louis University, Saint
Louis, Missouri 63103, United States
| | - Samuel Azibere
- Department
of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
| | - Mithila Tennakoon
- Department
of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
- Institute
for Drug and Biotherapeutic Innovation, Saint Louis University, Saint
Louis, Missouri 63103, United States
| | - John L. Payton
- College
of Sciences, University of Findlay, Findlay, Ohio 45840, United States
| | - Randy S. Sprague
- Department
of Pharmacology and Physiology, Saint Louis
University School of Medicine, Saint Louis, Missouri 63104, United States
| | - R. Scott Martin
- Department
of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
| | - Steven J. Sucheck
- Department
of Chemistry and Biochemistry, The University
of Toledo, Toledo, Ohio 43606, United States
| | - Ajith Karunarathne
- Department
of Chemistry, Saint Louis University, Saint Louis, Missouri 63103, United States
- Institute
for Drug and Biotherapeutic Innovation, Saint Louis University, Saint
Louis, Missouri 63103, United States
| |
Collapse
|
2
|
Sabirov RZ, Islam MR, Okada T, Merzlyak PG, Kurbannazarova RS, Tsiferova NA, Okada Y. The ATP-Releasing Maxi-Cl Channel: Its Identity, Molecular Partners and Physiological/Pathophysiological Implications. Life (Basel) 2021; 11:life11060509. [PMID: 34073084 PMCID: PMC8229958 DOI: 10.3390/life11060509] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/18/2022] Open
Abstract
The Maxi-Cl phenotype accounts for the majority (app. 60%) of reports on the large-conductance maxi-anion channels (MACs) and has been detected in almost every type of cell, including placenta, endothelium, lymphocyte, cardiac myocyte, neuron, and glial cells, and in cells originating from humans to frogs. A unitary conductance of 300-400 pS, linear current-to-voltage relationship, relatively high anion-to-cation selectivity, bell-shaped voltage dependency, and sensitivity to extracellular gadolinium are biophysical and pharmacological hallmarks of the Maxi-Cl channel. Its identification as a complex with SLCO2A1 as a core pore-forming component and two auxiliary regulatory proteins, annexin A2 and S100A10 (p11), explains the activation mechanism as Tyr23 dephosphorylation at ANXA2 in parallel with calcium binding at S100A10. In the resting state, SLCO2A1 functions as a prostaglandin transporter whereas upon activation it turns to an anion channel. As an efficient pathway for chloride, Maxi-Cl is implicated in a number of physiologically and pathophysiologically important processes, such as cell volume regulation, fluid secretion, apoptosis, and charge transfer. Maxi-Cl is permeable for ATP and other small signaling molecules serving as an electrogenic pathway in cell-to-cell signal transduction. Mutations at the SLCO2A1 gene cause inherited bone and gut pathologies and malignancies, signifying the Maxi-Cl channel as a perspective pharmacological target.
Collapse
Affiliation(s)
- Ravshan Z. Sabirov
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent 100174, Uzbekistan
- Correspondence: (R.Z.S.); (Y.O.); Tel.: +81-46-858-1501 (Y.O.); Fax: +81-46-858-1542 (Y.O.)
| | - Md. Rafiqul Islam
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Department of Biochemistry and Molecular Biology, Jagannath University, Dhaka 1100, Bangladesh
| | - Toshiaki Okada
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Veneno Technologies Co. Ltd., Tsukuba 305-0031, Japan
| | - Petr G. Merzlyak
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent 100174, Uzbekistan
| | - Ranokhon S. Kurbannazarova
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent 100174, Uzbekistan
| | - Nargiza A. Tsiferova
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent 100174, Uzbekistan
| | - Yasunobu Okada
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Department of Physiology, School of Medicine, Aichi Medical University, Nagakute 480-1195, Japan
- Correspondence: (R.Z.S.); (Y.O.); Tel.: +81-46-858-1501 (Y.O.); Fax: +81-46-858-1542 (Y.O.)
| |
Collapse
|
3
|
Vallon V, Unwin R, Inscho EW, Leipziger J, Kishore BK. Extracellular Nucleotides and P2 Receptors in Renal Function. Physiol Rev 2019; 100:211-269. [PMID: 31437091 DOI: 10.1152/physrev.00038.2018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The understanding of the nucleotide/P2 receptor system in the regulation of renal hemodynamics and transport function has grown exponentially over the last 20 yr. This review attempts to integrate the available data while also identifying areas of missing information. First, the determinants of nucleotide concentrations in the interstitial and tubular fluids of the kidney are described, including mechanisms of cellular release of nucleotides and their extracellular breakdown. Then the renal cell membrane expression of P2X and P2Y receptors is discussed in the context of their effects on renal vascular and tubular functions. Attention is paid to effects on the cortical vasculature and intraglomerular structures, autoregulation of renal blood flow, tubuloglomerular feedback, and the control of medullary blood flow. The role of the nucleotide/P2 receptor system in the autocrine/paracrine regulation of sodium and fluid transport in the tubular and collecting duct system is outlined together with its role in integrative sodium and fluid homeostasis and blood pressure control. The final section summarizes the rapidly growing evidence indicating a prominent role of the extracellular nucleotide/P2 receptor system in the pathophysiology of the kidney and aims to identify potential therapeutic opportunities, including hypertension, lithium-induced nephropathy, polycystic kidney disease, and kidney inflammation. We are only beginning to unravel the distinct physiological and pathophysiological influences of the extracellular nucleotide/P2 receptor system and the associated therapeutic perspectives.
Collapse
Affiliation(s)
- Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Robert Unwin
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Edward W Inscho
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Jens Leipziger
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Bellamkonda K Kishore
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| |
Collapse
|
4
|
Elorza-Vidal X, Gaitán-Peñas H, Estévez R. Chloride Channels in Astrocytes: Structure, Roles in Brain Homeostasis and Implications in Disease. Int J Mol Sci 2019; 20:ijms20051034. [PMID: 30818802 PMCID: PMC6429410 DOI: 10.3390/ijms20051034] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/15/2019] [Accepted: 02/17/2019] [Indexed: 12/29/2022] Open
Abstract
Astrocytes are the most abundant cell type in the CNS (central nervous system). They exert multiple functions during development and in the adult CNS that are essential for brain homeostasis. Both cation and anion channel activities have been identified in astrocytes and it is believed that they play key roles in astrocyte function. Whereas the proteins and the physiological roles assigned to cation channels are becoming very clear, the study of astrocytic chloride channels is in its early stages. In recent years, we have moved from the identification of chloride channel activities present in astrocyte primary culture to the identification of the proteins involved in these activities, the determination of their 3D structure and attempts to gain insights about their physiological role. Here, we review the recent findings related to the main chloride channels identified in astrocytes: the voltage-dependent ClC-2, the calcium-activated bestrophin, the volume-activated VRAC (volume-regulated anion channel) and the stress-activated Maxi-Cl−. We discuss key aspects of channel biophysics and structure with a focus on their role in glial physiology and human disease.
Collapse
Affiliation(s)
- Xabier Elorza-Vidal
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
- Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, 08907 Barcelona, Spain.
| | - Héctor Gaitán-Peñas
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
- Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, 08907 Barcelona, Spain.
| | - Raúl Estévez
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
- Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, 08907 Barcelona, Spain.
| |
Collapse
|
5
|
Okada Y, Okada T, Sato-Numata K, Islam MR, Ando-Akatsuka Y, Numata T, Kubo M, Shimizu T, Kurbannazarova RS, Marunaka Y, Sabirov RZ. Cell Volume-Activated and Volume-Correlated Anion Channels in Mammalian Cells: Their Biophysical, Molecular, and Pharmacological Properties. Pharmacol Rev 2019; 71:49-88. [PMID: 30573636 DOI: 10.1124/pr.118.015917] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
There are a number of mammalian anion channel types associated with cell volume changes. These channel types are classified into two groups: volume-activated anion channels (VAACs) and volume-correlated anion channels (VCACs). VAACs can be directly activated by cell swelling and include the volume-sensitive outwardly rectifying anion channel (VSOR), which is also called the volume-regulated anion channel; the maxi-anion channel (MAC or Maxi-Cl); and the voltage-gated anion channel, chloride channel (ClC)-2. VCACs can be facultatively implicated in, although not directly activated by, cell volume changes and include the cAMP-activated cystic fibrosis transmembrane conductance regulator (CFTR) anion channel, the Ca2+-activated Cl- channel (CaCC), and the acid-sensitive (or acid-stimulated) outwardly rectifying anion channel. This article describes the phenotypical properties and activation mechanisms of both groups of anion channels, including accumulating pieces of information on the basis of recent molecular understanding. To that end, this review also highlights the molecular identities of both anion channel groups; in addition to the molecular identities of ClC-2 and CFTR, those of CaCC, VSOR, and Maxi-Cl were recently identified by applying genome-wide approaches. In the last section of this review, the most up-to-date information on the pharmacological properties of both anion channel groups, especially their half-maximal inhibitory concentrations (IC50 values) and voltage-dependent blocking, is summarized particularly from the standpoint of pharmacological distinctions among them. Future physiologic and pharmacological studies are definitely warranted for therapeutic targeting of dysfunction of VAACs and VCACs.
Collapse
Affiliation(s)
- Yasunobu Okada
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Toshiaki Okada
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Kaori Sato-Numata
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Md Rafiqul Islam
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Yuhko Ando-Akatsuka
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Tomohiro Numata
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Machiko Kubo
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Takahiro Shimizu
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Ranohon S Kurbannazarova
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Yoshinori Marunaka
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Ravshan Z Sabirov
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| |
Collapse
|
6
|
Molecular Identities and ATP Release Activities of Two Types of Volume-Regulatory Anion Channels, VSOR and Maxi-Cl. CURRENT TOPICS IN MEMBRANES 2018; 81:125-176. [PMID: 30243431 DOI: 10.1016/bs.ctm.2018.07.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
An elaborate volume regulation system based on interplay of ion channels and transporters was evolved to cope with constant osmotic challenges caused by intensive metabolism, transport and other physiological/pathophysiological events. In animal cells, two types of anion channels are directly activated by cell swelling and involved in the regulatory volume decrease (RVD): volume-sensitive outwardly rectifying anion channel (VSOR), also called volume-regulated anion channel (VRAC), and Maxi-Cl which is the most major type of maxi-anion channel (MAC). These two channels have very different biophysical profiles and exhibit opposite dependence on intracellular ATP. After several decades of verifying many false-positive candidates for VSOR and Maxi-Cl, LRRC8 family proteins emerged as major VSOR components, and SLCO2A1 protein as a core of Maxi-Cl. Still, neither of these proteins alone can fully reproduce the native channel phenotypes suggesting existence of missing components. Although both VSOR and Maxi-Cl have pores wide enough to accommodate bulky ATP4- and MgATP2- anions, evidence accumulated hitherto, based on pharmacological and gene silencing experiments, suggests that Maxi-Cl, but not VSOR, serves as one of the major pathways for the release of ATP from swollen and ischemic/hypoxic cells. Relations of VSOR and Maxi-Cl with diseases and their selective pharmacology are the topics promoted by recent advance in molecular identification of the two volume-activated, volume-regulatory anion channels.
Collapse
|
7
|
Sabirov RZ, Merzlyak PG, Islam MR, Okada T, Okada Y. The properties, functions, and pathophysiology of maxi-anion channels. Pflugers Arch 2016; 468:405-20. [DOI: 10.1007/s00424-015-1774-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/11/2015] [Accepted: 12/15/2015] [Indexed: 01/19/2023]
|
8
|
Burow P, Klapperstück M, Markwardt F. Activation of ATP secretion via volume-regulated anion channels by sphingosine-1-phosphate in RAW macrophages. Pflugers Arch 2014; 467:1215-26. [PMID: 24965069 DOI: 10.1007/s00424-014-1561-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
We report the activation of outwardly rectifying anion currents by sphingosine-1-phosphate (S1P) in the murine macrophage cell line RAW 264.7. The S1P-induced current is mainly carried by anions, because the reversal potential of the current was shifted by replacement of extracellular Cl(-) by glutamate(-) but not when extracellular Na(+) was substituted by Tris(+). The inhibition of the current by hypertonic extracellular or hypotonic intracellular solution as well as the inhibitory effects of NPPB, tamoxifen, and glibenclamide indicates that the anion current is mediated by volume-regulated anion channels (VRAC). The S1P effect was blocked by intracellular GDPβS and W123, which points to signaling via the S1P receptor 1 (S1PR1) and G proteins. As cytochalasin D diminished the action of S1P, we conclude that the actin cytoskeleton is involved in the stimulation of VRAC. S1P and hypotonic extracellular solution induced secretion of ATP from the macrophages, which in both cases was blocked in a similar way by typical VRAC blockers. We suppose that the S1P-induced ATP secretion in macrophages via activation of VRAC constitutes a functional link between sphingolipid and purinergic signaling in essential processes such as inflammation and migration of leukocytes as well as phagocytosis and the killing of intracellular bacteria.
Collapse
Affiliation(s)
- Philipp Burow
- Julius Bernstein Institute for Physiology, Martin Luther University Halle, Magdeburger Str. 6, 06097, Halle/Saale, Germany
| | | | | |
Collapse
|
9
|
Skals M, Bjaelde RG, Reinholdt J, Poulsen K, Vad BS, Otzen DE, Leipziger J, Praetorius HA. Bacterial RTX toxins allow acute ATP release from human erythrocytes directly through the toxin pore. J Biol Chem 2014; 289:19098-109. [PMID: 24860098 DOI: 10.1074/jbc.m114.571414] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
ATP is as an extracellular signaling molecule able to amplify the cell lysis inflicted by certain bacterial toxins including the two RTX toxins α-hemolysin (HlyA) from Escherichia coli and leukotoxin A (LtxA) from Aggregatibacter actinomycetemcomitans. Inhibition of P2X receptors completely blocks the RTX toxin-induced hemolysis over a larger concentration range. It is, however, at present not known how the ATP that provides the amplification is released from the attacked cells. Here we show that both HlyA and LtxA trigger acute release of ATP from human erythrocytes that preceded and were not caused by cell lysis. This early ATP release did not occur via previously described ATP-release pathways in the erythrocyte. Both HlyA and LtxA were capable of triggering ATP release in the presence of the pannexin 1 blockers carbenoxolone and probenecid, and the HlyA-induced ATP release was found to be similar in erythrocytes from pannexin 1 wild type and knock-out mice. Moreover, the voltage-dependent anion channel antagonist TRO19622 had no effect on ATP release by either of the toxins. Finally, we showed that both HlyA and LtxA were able to release ATP from ATP-loaded lipid (1-palmitoyl-2-oleoyl-phosphatidylcholine) vesicles devoid of any erythrocyte channels or transporters. Again we were able to show that this happened in a non-lytic fashion, using calcein-containing vesicles as controls. These data show that both toxins incorporate into lipid vesicles and allow ATP to be released. We suggest that both toxins cause acute ATP release by letting ATP pass the toxin pores in both human erythrocytes and artificial membranes.
Collapse
Affiliation(s)
| | | | | | | | - Brian S Vad
- the Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center (iNANO), Center for Insoluble Proteins (inSPIN), Aarhus University, Aarhus C 8000, Denmark
| | - Daniel E Otzen
- the Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center (iNANO), Center for Insoluble Proteins (inSPIN), Aarhus University, Aarhus C 8000, Denmark
| | | | | |
Collapse
|
10
|
Shoji KF, Sáez PJ, Harcha PA, Aguila HL, Sáez JC. Pannexin1 channels act downstream of P2X 7 receptors in ATP-induced murine T-cell death. Channels (Austin) 2014; 8:142-56. [PMID: 24590064 DOI: 10.4161/chan.28122] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Death of murine T cells induced by extracellular ATP is mainly triggered by activation of purinergic P2X 7 receptors (P2X 7Rs). However, a link between P2X 7Rs and pannexin1 (Panx1) channels, which are non-selective, has been recently demonstrated in other cell types. In this work, we characterized the expression and cellular distribution of pannexin family members (Panxs 1, 2 and 3) in isolated T cells. Panx1 was the main pannexin family member clearly detected in both helper (CD4+) and cytotoxic (CD8+) T cells, whereas low levels of Panx2 were found in both T-cell subsets. Using pharmacological and genetic approaches, Panx1 channels were found to mediate most ATP-induced ethidium uptake since this was drastically reduced by Panx1 channel blockers (10Panx1, Probenecid and low carbenoxolone concentration) and absent in T cells derived from Panx1-/- mice. Moreover, electrophysiological measurements in wild-type CD4+ cells treated with ATP unitary current events and pharmacological sensitivity compatible with Panx1 channels were found. In addition, ATP release from T cells treated with 4Br-A23187, a calcium ionophore, was completely blocked with inhibitors of both connexin hemichannels and Panx1 channels. Panx1 channel blockers drastically reduced the ATP-induced T-cell mortality, indicating that Panx1 channels mediate the ATP-induced T-cell death. However, mortality was not reduced in T cells of Panx1-/- mice, in which levels of P2X 7Rs and ATP-induced intracellular free Ca2+ responses were enhanced suggesting that P2X 7Rs take over Panx1 channels lose-function in mediating the onset of cell death induced by extracellular ATP.
Collapse
Affiliation(s)
- Kenji F Shoji
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Pablo J Sáez
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Paloma A Harcha
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Hector L Aguila
- Department of Immunology; University of Connecticut Health Center; Farmington, CT USA
| | - Juan C Sáez
- Departamento de Fisiología; Pontificia Universidad Católica de Chile; Santiago, Chile; Instituto Milenio; Centro Interdisciplinario de Neurociencias de Valparaíso; Valparaíso, Chile
| |
Collapse
|
11
|
Islam MR, Uramoto H, Okada T, Sabirov RZ, Okada Y. Maxi-anion channel and pannexin 1 hemichannel constitute separate pathways for swelling-induced ATP release in murine L929 fibrosarcoma cells. Am J Physiol Cell Physiol 2012; 303:C924-35. [PMID: 22785119 DOI: 10.1152/ajpcell.00459.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The maxi-anion channel plays a classically recognized role in controlling the membrane potential through the chloride conductance. It also has novel functions as a regulated pathway for the release of the anionic signaling molecules ATP and excitatory amino acids from cells subjected to osmotic perturbation, ischemia, or hypoxia. Because hemichannels formed by pannexins and connexins have been reported to mediate ATP release from a number of cell types, these hemichannels may represent the molecular correlate of the maxi-anion channel. Here, we found that L929 fibrosarcoma cells express functional maxi-anion channels which mediate a major portion of swelling-induced ATP release, and that ATP released via maxi-anion channels facilitates the regulatory volume decrease after osmotic swelling. Also, it was found that the cells express the mRNA for pannexin 1, pannexin 2, and connexin 43. Hypotonicity-induced ATP release was partially suppressed not only by known blockers of the maxi-anion channel but also by several blockers of pannexins including the pannexin 1-specific blocking peptide (10)Panx1 and small interfering (si)RNA against pannexin 1 but not pannexin 2. The inhibitory effects of maxi-anion channel blockers and pannexin 1 antagonists were additive. In contrast, maxi-anion channel activity was not affected by pannexin 1 antagonists and siRNAs against pannexins 1 and 2. Although a connexin 43-specific blocking peptide, Gap27, slightly suppressed hypotonicity-induced ATP release, maxi-anion channel activity was not affected by Gap27 or connexin 43-specific siRNA. Thus, it is concluded that the maxi-anion channel is a molecular entity distinct from pannexin 1, pannexin 2, and connexin 43, and that the maxi-anion channel and the hemichannels constitute separate pathways for swelling-induced ATP release in L929 cells.
Collapse
Affiliation(s)
- Md Rafiqul Islam
- Dept. of Cell Physiology, National Institute for Physiological Sciences, Myodaiji-cho, Okazaki 444-8585, Japan
| | | | | | | | | |
Collapse
|
12
|
Birder LA, Ruggieri M, Takeda M, van Koeveringe G, Veltkamp S, Korstanje C, Parsons B, Fry CH. How does the urothelium affect bladder function in health and disease? ICI-RS 2011. Neurourol Urodyn 2012; 31:293-9. [PMID: 22275289 DOI: 10.1002/nau.22195] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 12/06/2011] [Indexed: 11/11/2022]
Abstract
The urothelium is a multifunctional tissue that not only acts as a barrier between the vesical contents of the lower urinary tract and the underlying tissues but also acts as a sensory organ by transducing physical and chemical stresses to the attendant afferent nervous system and underlying smooth muscle. This review will consider the nature of the stresses that the urothelium can transduce; the transmitters that mediate the transduction process; and how lower urinary pathologies, including overactive bladder syndrome, painful bladder syndrome and bacterial infections, are associated with alterations to this sensory system. In particular, the role of muscarinic receptors and the TRPV channels system will be discussed in this context. The urothelium also influences the contractile state of detrusor smooth muscle, both through modifying its contractility and the extent of spontaneous activity; potential pathways are discussed. The potential role that the urothelium may play in bladder underactivity is introduced, as well as potential biomarkers for the condition that may cross the urothelium to the urine. Finally, consideration is given to vesical administration of therapeutic agents that influence urinary tract function and how the properties of the urothelium may determine the effectiveness of this mode of delivery.
Collapse
Affiliation(s)
- L A Birder
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Swelling-activated anion channels are essential for volume regulation of mouse thymocytes. Int J Mol Sci 2011; 12:9125-37. [PMID: 22272123 PMCID: PMC3257120 DOI: 10.3390/ijms12129125] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 11/10/2011] [Accepted: 11/24/2011] [Indexed: 11/16/2022] Open
Abstract
Channel-mediated trans-membrane chloride movement is a key process in the active cell volume regulation under osmotic stress in most cells. However, thymocytes were hypothesized to regulate their volume by activating a coupled K-Cl cotransport mechanism. Under the patch-clamp, we found that osmotic swelling activates two types of macroscopic anion conductance with different voltage-dependence and pharmacology. At the single-channel level, we identified two types of events: one corresponded to the maxi-anion channel, and the other one had characteristics of the volume-sensitive outwardly rectifying (VSOR) chloride channel of intermediate conductance. A VSOR inhibitor, phloretin, significantly suppressed both macroscopic VSOR-type conductance and single-channel activity of intermediate amplitude. The maxi-anion channel activity was largely suppressed by Gd(3+) ions but not by phloretin. Surprisingly, [(dihydroindenyl)oxy] alkanoic acid (DIOA), a known antagonist of K-Cl cotransporter, was found to significantly suppress the activity of the VSOR-type single-channel events with no effect on the maxi-anion channels at 10 μM. The regulatory volume decrease (RVD) phase of cellular response to hypotonicity was mildly suppressed by Gd(3+) ions and was completely abolished by phloretin suggesting a major impact of the VSOR chloride channel and modulatory role of the maxi-anion channel. The inhibitory effect of DIOA was also strong, and, most likely, it occurred via blocking the VSOR Cl(-) channels.
Collapse
|
14
|
Sabirov RZ, Merzlyak PG. Plasmalemmal VDAC controversies and maxi-anion channel puzzle. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:1570-80. [PMID: 21986486 DOI: 10.1016/j.bbamem.2011.09.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 09/22/2011] [Accepted: 09/23/2011] [Indexed: 12/14/2022]
Abstract
The maxi-anion channel has been observed in many cell types from the very beginning of the patch-clamp era. The channel is highly conductive for chloride and thus can modulate the resting membrane potential and play a role in fluid secretion/absorption and cell volume regulation. A wide nanoscopic pore of the maxi-anion channel permits passage of excitatory amino acids and nucleotides. The channel-mediated release of these signaling molecules is associated with kidney tubuloglomerular feedback, cardiac ischemia/hypoxia, as well as brain ischemia/hypoxia and excitotoxic neurodegeneration. Despite the ubiquitous expression and physiological/pathophysiological significance, the molecular identity of the maxi-anion channel is still obscure. VDAC is primarily a mitochondrial protein; however several groups detected it on the cellular surface. VDAC in lipid bilayers reproduced the most important biophysical properties of the maxi-anion channel, such as a wide nano-sized pore, closure in response to moderately high voltages, ATP-block and ATP-permeability. However, these similarities turned out to be superficial, and the hypothesis of plasmalemmal VDAC as the maxi-anion channel did not withstand the test by genetic manipulations of VDAC protein expression. VDAC on the cellular surface could also function as a ferricyanide reductase or a receptor for plasminogen kringle 5 and for neuroactive steroids. These ideas, as well as the very presence of VDAC on plasmalemma, remain to be scrutinized by genetic manipulations of the VDAC protein expression. This article is part of a Special Issue entitled: VDAC structure, function, and regulation of mitochondrial metabolism.
Collapse
Affiliation(s)
- Ravshan Z Sabirov
- Laboratory of Molecular Physiology, Institute of Pysiology and Biphysics, Academy of Science, RUz, Tashkent, Uzbekistan.
| | | |
Collapse
|
15
|
TRP channels in urinary bladder mechanosensation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:861-79. [PMID: 21290331 DOI: 10.1007/978-94-007-0265-3_45] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Vallon V, Rieg T. Regulation of renal NaCl and water transport by the ATP/UTP/P2Y2 receptor system. Am J Physiol Renal Physiol 2011; 301:F463-75. [PMID: 21715471 DOI: 10.1152/ajprenal.00236.2011] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extracellular nucleotides (e.g., ATP) activate ionotropic P2X and metabotropic P2Y receptors in the plasma membrane to regulate and maintain cell function and integrity. This includes the renal tubular and collecting duct system, where the locally released nucleotides act in a paracrine and autocrine way to regulate transport of electrolytes and water and maintain cell volume. A prominent role has been assigned to Gq-coupled P2Y(2) receptors, which are typically activated by both ATP and UTP. Studies in gene knockout mice revealed an antihypertensive activity of P2Y(2) receptors that is linked to vasodilation and an inhibitory influence on renal salt reabsorption. Flow induces apical ATP release in the thick ascending limb, and first evidence indicates an inhibitory influence of P2Y(2) receptor tone on the expression and activity of the Na-K-2Cl cotransporter NKCC2 in this segment. The apical ATP/UTP/P2Y(2) receptor system in the connecting tubule/cortical collecting duct mediates the inhibitory effect of dietary salt on the open probability of the epithelial sodium channel ENaC and inhibits ENaC activity during aldosterone escape. Connexin 30 has been implicated in the luminal release of the ATP involved in the regulation of ENaC. An increase in collecting duct cell volume in response to manipulating water homeostasis increases ATP release. The subsequent activation of P2Y(2) receptors inhibits vasopressin-induced cAMP formation and water reabsorption, which facilitates water excretion and stabilizes cell volume. Thus recent studies have established the ATP/UTP/P2Y(2) receptor system as a relevant regulator of renal salt and water homeostasis and blood pressure regulation. The pathophysiological relevance and therapeutic potential remains to be determined, but dual effects of P2Y(2) receptor activation on both the vasculature and renal salt reabsorption implicate these receptors as potential therapeutic targets in hypertension.
Collapse
Affiliation(s)
- Volker Vallon
- Dept. of Medicine, Univ. of California San Diego, 92161, USA.
| | | |
Collapse
|
17
|
Araki I, Yoshiyama M, Kobayashi H, Mochizuki T, Du S, Okada Y, Takeda M. Emerging Families of Ion Channels Involved in Urinary Bladder Nociception. Pharmaceuticals (Basel) 2010; 3:2248-2267. [PMID: 27713353 PMCID: PMC4036652 DOI: 10.3390/ph3072248] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 06/28/2010] [Accepted: 07/15/2010] [Indexed: 01/12/2023] Open
Abstract
The expression of multiple ion channels and receptors is essential for nociceptors to detect noxious stimuli of a thermal, mechanical or chemical nature. The peripheral sensory transduction systems of the urinary bladder include sensory nerve endings, urothelial cells and others whose location is suitable for transducing mechanical and chemical stimuli. There is an increasing body of evidence implicating the Deg/ENaC and TRP channel families in the control of bladder afferent excitability under physiological and pathological conditions. Pharmacological interventions targeting these ion channels may provide a new strategy for the treatment of pathological bladder sensation and pain.
Collapse
Affiliation(s)
- Isao Araki
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Mitsuharu Yoshiyama
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
| | - Hideki Kobayashi
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
| | - Tsutomu Mochizuki
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
| | - Shuqi Du
- Department of Urology, the 1st Affiliated Hospital, China Medical University, Shenyang, China.
| | - Yusaku Okada
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Masayuki Takeda
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
| |
Collapse
|
18
|
Sridharan M, Adderley SP, Bowles EA, Egan TM, Stephenson AH, Ellsworth ML, Sprague RS. Pannexin 1 is the conduit for low oxygen tension-induced ATP release from human erythrocytes. Am J Physiol Heart Circ Physiol 2010; 299:H1146-52. [PMID: 20622111 DOI: 10.1152/ajpheart.00301.2010] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Erythrocytes release ATP in response to exposure to the physiological stimulus of lowered oxygen (O(2)) tension as well as pharmacological activation of the prostacyclin receptor (IPR). ATP release in response to these stimuli requires activation of adenylyl cyclase, accumulation of cAMP, and activation of protein kinase A. The mechanism by which ATP, a highly charged anion, exits the erythrocyte in response to lowered O(2) tension or receptor-mediated IPR activation by iloprost is unknown. It was demonstrated previously that inhibiting pannexin 1 with carbenoxolone inhibits hypotonically induced ATP release from human erythrocytes. Here we demonstrate that three structurally dissimilar compounds known to inhibit pannexin 1 prevent ATP release in response to lowered O(2) tension but not to iloprost-induced ATP release. These results suggest that pannexin 1 is the conduit for ATP release from erythrocytes in response to lowered O(2) tension. However, the identity of the conduit for iloprost-induced ATP release remains unknown.
Collapse
Affiliation(s)
- Meera Sridharan
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
TAKEDA M, MOCHIZUKI T, YOSHIYAMA M, NAKAGOMI H, KOBAYASHI H, SAWADA N, ZAKOHJI H, DU S, ARAKI I. Sensor Mechanism and Afferent Signal Transduction of the Urinary Bladder: Special Focus on transient receptor potential Ion Channels. Low Urin Tract Symptoms 2010; 2:51-60. [DOI: 10.1111/j.1757-5672.2010.00074.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
20
|
Hua SZ, Gottlieb PA, Heo J, Sachs F. A mechanosensitive ion channel regulating cell volume. Am J Physiol Cell Physiol 2010; 298:C1424-30. [PMID: 20457830 DOI: 10.1152/ajpcell.00503.2009] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Cells respond to a hyposmotic challenge by swelling and then returning toward the resting volume, a process known as the regulatory volume decrease or RVD. The sensors for this process have been proposed to include cationic mechanosensitive ion channels that are opened by membrane tension. We tested this hypothesis using a microfluidic device to measure cell volume and the peptide GsMTx4, a specific inhibitor of cationic mechanosensitive channels. GsMTx4 had no effect on RVD in primary rat astrocytes or Madin-Darby canine kidney (MDCK) cells but was able to completely inhibit RVD and the associated Ca(2+) uptake in normal rat kidney (NRK-49F) cells in a dose-dependent manner. Gadolinium (Gd(3+)), a nonspecific blocker of many mechanosensitive channels, inhibited RVD and Ca(2+) uptake in all three cell types, demonstrating the existence of at least two types of volume sensors. Single-channel stretch-activated currents are present in outside-out patches from NRK-49F, MDCK, and astrocytes, and they are reversibly inhibited by GsMTx4. While mechanosensitive channels are involved in volume regulation, their role for volume sensing is specialized. The NRK cells form a stable platform from which to screen drugs that affect volume regulation via mechanosensory channels and as a sensitive system to clone the channel.
Collapse
Affiliation(s)
- Susan Z Hua
- Department of Physiology and Biophysics, SUNY, Buffalo, NY 14214, USA.
| | | | | | | |
Collapse
|
21
|
Toychiev AH, Sabirov RZ, Takahashi N, Ando-Akatsuka Y, Liu H, Shintani T, Noda M, Okada Y. Activation of maxi-anion channel by protein tyrosine dephosphorylation. Am J Physiol Cell Physiol 2009; 297:C990-1000. [PMID: 19657061 DOI: 10.1152/ajpcell.00131.2009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The maxi-anion channel with a large single-channel conductance of >300 pS, and unknown molecular identity, is functionally expressed in a large variety of cell types. The channel is activated by a number of experimental maneuvers such as exposing cells to hypotonic or ischemic stress. The most effective and consistent method of activating it is patch membrane excision. However, the activation mechanism of the maxi-anion channel remains poorly understood at present. In the present study, involvement of phosphorylation/dephosphorylation in excision-induced activation was examined. In mouse mammary fibroblastic C127 cells, activity of the channel was suppressed by intracellular application of Mg-ATP, but not Mg-5'-adenylylimidodiphosphate (AMP-PNP), in a concentration-dependent manner. When a cocktail of broad-spectrum tyrosine phosphatase inhibitors was applied, channel activation was completely abolished, whereas inhibitors of serine/threonine protein phosphatases had no effect. On the other hand, protein tyrosine kinase inhibitors brought the channel out of an inactivated state. In mouse adult skin fibroblasts (MAFs) in primary culture, similar maxi-anion channels were found to be activated on membrane excision, in a manner sensitive to tyrosine phosphatase inhibitors. In MAFs isolated from animals deficient in receptor protein tyrosine phosphatase (RPTP)zeta, activation of the maxi-anion channel was significantly slower and less prominent compared with that observed in wild-type MAFs; however, channel activation was restored by transfection of the RPTPzeta gene. Thus it is concluded that activation of the maxi-anion channel involves protein dephosphorylation mediated by protein tyrosine phosphatases that include RPTPzeta in mouse fibroblasts, but not in C127 cells.
Collapse
Affiliation(s)
- Abduqodir H Toychiev
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Mizumori M, Ham M, Guth PH, Engel E, Kaunitz JD, Akiba Y. Intestinal alkaline phosphatase regulates protective surface microclimate pH in rat duodenum. J Physiol 2009; 587:3651-63. [PMID: 19451200 DOI: 10.1113/jphysiol.2009.172270] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Regulation of localized extracellular pH (pH(o)) maintains normal organ function. An alkaline microclimate overlying the duodenal enterocyte brush border protects the mucosa from luminal acid. We hypothesized that intestinal alkaline phosphatase (IAP) regulates pH(o) due to pH-sensitive ATP hydrolysis as part of an ecto-purinergic pH regulatory system, comprised of cell-surface P2Y receptors and ATP-stimulated duodenal bicarbonate secretion (DBS). To test this hypothesis, we measured DBS in a perfused rat duodenal loop, examining the effect of the competitive alkaline phosphatase inhibitor glycerol phosphate (GP), the ecto-nucleoside triphosphate diphosphohydrolase inhibitor ARL67156, and exogenous nucleotides or P2 receptor agonists on DBS. Furthermore, we measured perfusate ATP concentration with a luciferin-luciferase bioassay. IAP inhibition increased DBS and luminal ATP output. Increased luminal ATP output was partially CFTR dependent, but was not due to cellular injury. Immunofluorescence localized the P2Y(1) receptor to the brush border membrane of duodenal villi. The P2Y(1) agonist 2-methylthio-ADP increased DBS, whereas the P2Y(1) antagonist MRS2179 reduced ATP- or GP-induced DBS. Acid perfusion augmented DBS and ATP release, further enhanced by the IAP inhibitor l-cysteine, and reduced by the exogenous ATPase apyrase. Furthermore, MRS2179 or the highly selective P2Y(1) antagonist MRS2500 co-perfused with acid induced epithelial injury, suggesting that IAP/ATP/P2Y signalling protects the mucosa from acid injury. Increased DBS augments IAP activity presumably by raising pH(o), increasing the rate of ATP degradation, decreasing ATP-mediated DBS, forming a negative feedback loop. The duodenal epithelial brush border IAP-P2Y-HCO(3-) surface microclimate pH regulatory system effectively protects the mucosa from acid injury.
Collapse
Affiliation(s)
- Misa Mizumori
- Department of Medicine, School of Medicine, University of California Los Angeles, CA 90073, USA
| | | | | | | | | | | |
Collapse
|
23
|
Hoffmann EK, Lambert IH, Pedersen SF. Physiology of cell volume regulation in vertebrates. Physiol Rev 2009; 89:193-277. [PMID: 19126758 DOI: 10.1152/physrev.00037.2007] [Citation(s) in RCA: 1054] [Impact Index Per Article: 65.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The ability to control cell volume is pivotal for cell function. Cell volume perturbation elicits a wide array of signaling events, leading to protective (e.g., cytoskeletal rearrangement) and adaptive (e.g., altered expression of osmolyte transporters and heat shock proteins) measures and, in most cases, activation of volume regulatory osmolyte transport. After acute swelling, cell volume is regulated by the process of regulatory volume decrease (RVD), which involves the activation of KCl cotransport and of channels mediating K(+), Cl(-), and taurine efflux. Conversely, after acute shrinkage, cell volume is regulated by the process of regulatory volume increase (RVI), which is mediated primarily by Na(+)/H(+) exchange, Na(+)-K(+)-2Cl(-) cotransport, and Na(+) channels. Here, we review in detail the current knowledge regarding the molecular identity of these transport pathways and their regulation by, e.g., membrane deformation, ionic strength, Ca(2+), protein kinases and phosphatases, cytoskeletal elements, GTP binding proteins, lipid mediators, and reactive oxygen species, upon changes in cell volume. We also discuss the nature of the upstream elements in volume sensing in vertebrate organisms. Importantly, cell volume impacts on a wide array of physiological processes, including transepithelial transport; cell migration, proliferation, and death; and changes in cell volume function as specific signals regulating these processes. A discussion of this issue concludes the review.
Collapse
Affiliation(s)
- Else K Hoffmann
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | | | | |
Collapse
|
24
|
Sabirov RZ, Okada Y. The maxi-anion channel: a classical channel playing novel roles through an unidentified molecular entity. J Physiol Sci 2009; 59:3-21. [PMID: 19340557 PMCID: PMC10717152 DOI: 10.1007/s12576-008-0008-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Accepted: 11/05/2008] [Indexed: 10/20/2022]
Abstract
The maxi-anion channel is widely expressed and found in almost every part of the body. The channel is activated in response to osmotic cell swelling, to excision of the membrane patch, and also to some other physiologically and pathophysiologically relevant stimuli, such as salt stress in kidney macula densa as well as ischemia/hypoxia in heart and brain. Biophysically, the maxi-anion channel is characterized by a large single-channel conductance of 300-400 pS, which saturates at 580-640 pS with increasing the Cl(-) concentration. The channel discriminates well between Na(+) and Cl(-), but is poorly selective to other halides exhibiting weak electric-field selectivity with an Eisenman's selectivity sequence I. The maxi-anion channel has a wide pore with an effective radius of approximately 1.3 nm and permits passage not only of Cl(-) but also of some intracellular large organic anions, thereby releasing major extracellular signals and gliotransmitters such as glutamate(-) and ATP(4-). The channel-mediated efflux of these signaling molecules is associated with kidney tubuloglomerular feedback, cardiac ischemia/hypoxia, as well as brain ischemia/hypoxia and excitotoxic neurodegeneration. Despite the ubiquitous expression, well-defined properties and physiological/pathophysiological significance of this classical channel, the molecular entity has not been identified. Molecular identification of the maxi-anion channel is an urgent task that would greatly promote investigation in the fields not only of anion channel but also of physiological/pathophysiological signaling in the brain, heart and kidney.
Collapse
Affiliation(s)
- Ravshan Z. Sabirov
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585 Japan
- Laboratory of Molecular Physiology, Institute of Physiology and Biophysics, Tashkent, 100095 Uzbekistan
| | - Yasunobu Okada
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585 Japan
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (Sokendai), Okazaki, 444-8585 Japan
| |
Collapse
|
25
|
Abstract
Chemotaxis allows polymorphonuclear neutrophils (PMN) to rapidly reach infected and inflamed sites. However, excessive influx of PMN damages host tissues. Better knowledge of the mechanisms that control PMN chemotaxis may lead to improved treatments of inflammatory diseases. Recent findings suggest that ATP and adenosine are involved in PMN chemotaxis. Therefore, these purinergic signaling processes may be suitable targets for novel therapeutic approaches to ameliorate host tissue damage.
Collapse
Affiliation(s)
- W G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, East Stoneman 8M 10C, Boston, Massachusetts 02215, USA.
| |
Collapse
|
26
|
Franco R, Panayiotidis MI, de la Paz LDO. Autocrine signaling involved in cell volume regulation: the role of released transmitters and plasma membrane receptors. J Cell Physiol 2008; 216:14-28. [PMID: 18300263 DOI: 10.1002/jcp.21406] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cell volume regulation is a basic homeostatic mechanism transcendental for the normal physiology and function of cells. It is mediated principally by the activation of osmolyte transport pathways that result in net changes in solute concentration that counteract cell volume challenges in its constancy. This process has been described to be regulated by a complex assortment of intracellular signal transduction cascades. Recently, several studies have demonstrated that alterations in cell volume induce the release of a wide variety of transmitters including hormones, ATP and neurotransmitters, which have been proposed to act as extracellular signals that regulate the activation of cell volume regulatory mechanisms. In addition, changes in cell volume have also been reported to activate plasma membrane receptors (including tyrosine kinase receptors, G-protein coupled receptors and integrins) that have been demonstrated to participate in the regulatory process of cell volume. In this review, we summarize recent studies about the role of changes in cell volume in the regulation of transmitter release as well as in the activation of plasma membrane receptors and their further implications in the regulation of the signaling machinery that regulates the activation of osmolyte flux pathways. We propose that the autocrine regulation of Ca2+-dependent and tyrosine phosphorylation-dependent signaling pathways by the activation of plasma membrane receptors and swelling-induced transmitter release is necessary for the activation/regulation of osmolyte efflux pathways and cell volume recovery. Furthermore, we emphasize the importance of studying these extrinsic signals because of their significance in the understanding of the physiology of cell volume regulation and its role in cell biology in vivo, where the constraint of the extracellular space might enhance the autocrine or even paracrine signaling induced by these released transmitters.
Collapse
Affiliation(s)
- Rodrigo Franco
- Laboratory of Cell Biology and Signal Transduction, Biomedical Research Unit, FES-Iztacala, UNAM, Mexico.
| | | | | |
Collapse
|
27
|
Araki I, Du S, Kobayashi H, Sawada N, Mochizuki T, Zakoji H, Takeda M. Roles of mechanosensitive ion channels in bladder sensory transduction and overactive bladder. Int J Urol 2008; 15:681-7. [PMID: 18462357 DOI: 10.1111/j.1442-2042.2008.02052.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In the storage phase, mechanical stretch stimulates bladder afferents. These signals generate sensations and trigger voiding responses, however the precise mechanisms by which mechanical stimuli excite bladder afferents are yet to be explored. For mechanosensory transduction, the presence of mechanosensors is essential in the peripheral sensory systems including sensory nerve endings, urothelium and others. There is increasing evidence that mechanosensitive ion channels, such as degenerin/epithelial Na(+) channel (ENaC) and transient receptor potential (TRP) channel families, play key roles in the mechanosensory transduction of the urinary bladder. Pharmacological interventions targeting mechanosensitive ion channels may provide a new strategy for the treatment of bladder dysfunction.
Collapse
Affiliation(s)
- Isao Araki
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Japan.
| | | | | | | | | | | | | |
Collapse
|
28
|
Lang F, Gulbins E, Szabo I, Vereninov A, Huber SM. Ion Channels, Cell Volume, Cell Proliferation and Apoptotic Cell Death. SENSING WITH ION CHANNELS 2008. [DOI: 10.1007/978-3-540-72739-2_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
29
|
Abstract
Survival of human and animal cells requires avoidance of excessive alterations of cell volume. The osmolarity amassed by cellular accumulation of organic substances must be compensated by lowering cytosolic ion concentrations. The Na+/K+ ATPase extrudes Na+ in exchange for K+, which can permeate the cell membrane through K+ channels. K+ exit generates a cell-negative potential difference across the cell membrane, driving the exit of anions such as Cl-. The low cytosolic Cl- concentrations counterbalance the excess cellular osmolarity by organic substances. Cell volume regulation following cell swelling involves releasing ions through activation of K+ channels and/or anion channels, KCl-cotransport, or parallel activation of K+/H+ exchange and Cl-/HCO3- exchange. Cell volume regulation following cell shrinkage involves accumulation of ions through activation of Na+,K+,2Cl- cotransport, Na+/H+ exchange in parallel to Cl-/HCO3- exchange, or Na+ channels. The Na+ taken up is extruded by the Na+/K+ ATPase in exchange for K+. Shrunken cells further accumulate organic osmolytes such as sorbitol and glycerophosphorylcholine, and monomeric amino acids by altered metabolism and myoinositol (inositol), betaine, taurine, and amino acids by Na+ coupled transport. They release osmolytes during cell swelling. Challenges of cell volume homeostasis include transport, hormones, transmitters, and drugs. Moreover, alterations of cell volume participate in the machinery regulating cell proliferation and apoptotic cell death. Deranged cell volume regulation significantly contributes to the pathophysiology of several disorders such as liver insufficiency, diabetic ketoacidosis, hypercatabolism, fibrosing disease, sickle cell anemia, and infection.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology I, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
30
|
Abstract
Connexins form intercellular channels that span two plasma membranes and directly couple the cytoplasm of adjacent cells. This morphological contact enables the exchange of ions, second messengers, and metabolites, which act to regulate several biological functions. This review focuses on the significance of connexins in the renal circulation. Cells of the renal vasculature are coupled and express connexins in a vessel and cell-specific pattern. This finding indicates that renal connexins likely play an important role in renal autoregulatory mechanisms (Bayliss effect, tubuloglomerular feedback) and in the control of vasomotor responses. The described coupling of endothelial and vascular smooth muscle cells in the afferent arterioles may also contribute to the communication of neighboring nephrons, called 'nephron coupling.' Furthermore, deletion of the Cx40 and Cx43 genes results in an altered functional behavior of the renin-producing cells, suggesting involvement of these connexin isoforms in the regulation of renin secretion and synthesis. In addition, this review discusses the role of renal connexin expression in the pathogenesis of hypertension or diabetes.
Collapse
Affiliation(s)
- C Wagner
- Physiologisches Institut der Universität Regensburg, Regensburg, Germany.
| |
Collapse
|
31
|
Spatial distribution of maxi-anion channel on cardiomyocytes detected by smart-patch technique. Biophys J 2007; 94:1646-55. [PMID: 18024498 DOI: 10.1529/biophysj.107.117820] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Spatial distribution of maxi-anion channels in rat cardiomyocytes were studied by applying the recently developed patch clamp technique under scanning ion conductance microscopy, called the "smart-patch" technique. In primary-cultured neonatal cells, the channel was found to be unevenly distributed over the cell surface with significantly lower channel activity in cellular extensions compared with the other parts. Local ATP release, detected using a PC12 cell-based biosensor technique, also exhibited a similar pattern. The maxi-anion channel activity could not be detected in freshly isolated adult cardiomyocytes by the conventional patch-clamp with 2-MOmega pipettes. However, when fine-tipped 15-20 MOmega pipettes were targeted to only Z-line areas, we observed, for the first time, the maxi-anion events. Smart-patching different regions of the cell surface, we found that the channel activity was maximal at the openings of T-tubules and along Z-lines, but was significantly decreased in the scallop crest area. Thus, it is concluded that maxi-anion channels are concentrated at the openings of T-tubules and along Z-lines in adult cardiomyocytes. This study showed that the smart-patch technique provides a powerful method to detect a unitary event of channels that are localized at some specific site in the narrow region.
Collapse
|
32
|
Vallon V. P2 receptors in the regulation of renal transport mechanisms. Am J Physiol Renal Physiol 2007; 294:F10-27. [PMID: 17977905 DOI: 10.1152/ajprenal.00432.2007] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Extracellular nucleotides (e.g., ATP) regulate physiological and pathophysiological processes through activation of nucleotide P2 receptors in the plasma membrane. Examples include such diverse processes as communication from taste buds to gustatory nerves, platelet aggregation, nociception, or neutrophil chemotaxis. Over approximately the last 15 years, evidence has also accumulated that cells in renal epithelia release nucleotides in response to physiological stimuli and that these nucleotides act in a paracrine and autocrine way to activate P2 receptors and play a significant role in the regulation of transport mechanisms and cell volume regulation. This review discusses potential stimuli and mechanisms involved in nucleotide release in renal epithelia and summarizes the available data on the expression and function of nucleotide P2 receptors along the native mammalian tubular and collecting duct system. Using established agonist profiles for P2 receptor subtypes, significant insights have been gained particularly into a potential role for P2Y(2)-like receptors in the regulation of transport mechanisms in the collecting duct. Due to the lack of receptor subtype-specific antagonists, however, the in vivo relevance of P2 receptor subtypes is unclear. Studies in gene knockout mice provided first insights including an antihypertensive activity of P2Y(2) receptors that is linked to an inhibitory influence on renal Na(+) and water reabsorption. We are only beginning to unravel the important roles of extracellular nucleotides and P2 receptors in the regulation of the diverse transport mechanisms of the kidney.
Collapse
Affiliation(s)
- Volker Vallon
- Department of Medicine, University of California and Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Dr., San Diego, CA 92161., USA.
| |
Collapse
|
33
|
Rieg T, Bundey RA, Chen Y, Deschenes G, Junger W, Insel PA, Vallon V. Mice lacking P2Y2 receptors have salt-resistant hypertension and facilitated renal Na+ and water reabsorption. FASEB J 2007; 21:3717-26. [PMID: 17575258 DOI: 10.1096/fj.07-8807com] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Extracellular nucleotides (e.g., ATP) regulate many physiological and pathophysiological processes through activation of nucleotide (P2) receptors in the plasma membrane. Here we report that gene-targeted (knockout) mice that lack P2Y2 receptors have salt-resistant arterial hypertension in association with an inverse relationship between salt intake and heart rate, indicating intact baroreceptor function. Knockout mice have multiple alterations in their handling of salt and water: these include suppressed plasma renin and aldosterone concentrations, lower renal expression of the aldosterone-induced epithelial sodium channel alpha-ENaC, greater medullary expression of the Na-K-2Cl-cotransporter NKCC2, and greater furosemide-sensitive Na+ reabsorption in association with greater renal medullary expression of aquaporin-2 and vasopressin-dependent renal cAMP formation and water reabsorption despite similar vasopressin levels compared with wild type. Of note, smaller increases in plasma aldosterone were required to adapt renal Na+ excretion to restricted intake in knockout mice, suggesting a facilitation in renal Na+ retention. The results thus identify a previously unrecognized role for P2Y2 receptors in blood pressure regulation that is linked to an inhibitory influence on renal Na+ and water reabsorption. Based on these findings in knockout mice, we propose that a blunting in P2Y2 receptor expression or activity is a new mechanism for salt-resistant arterial hypertension.
Collapse
Affiliation(s)
- Timo Rieg
- Department of Medicine, University of California San Diego, San Diego, CA 92161, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Vertebrates express two families of gap junction proteins: the well characterized connexins and the recently discovered pannexins. The latter are related to invertebrate innexins. Here we present the hypothesis that pannexins, rather than providing a redundant system to gap junctions formed by connexins, exert a physiological role as nonjunctional membrane channels. Specifically, we propose that pannexins can serve as ATP release channels. This function presumptively is also performed by innexins in invertebrates, in addition to their traditional gap junction role.
Collapse
Affiliation(s)
- Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami, School of Medicine, Miami, Florida 33101, USA.
| | | |
Collapse
|
35
|
Wang HY, Shimizu T, Numata T, Okada Y. Role of acid-sensitive outwardly rectifying anion channels in acidosis-induced cell death in human epithelial cells. Pflugers Arch 2006; 454:223-33. [PMID: 17186306 DOI: 10.1007/s00424-006-0193-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Accepted: 11/20/2006] [Indexed: 10/23/2022]
Abstract
Recently, a novel type of anion channel activated by extracellular acidification has been found in a variety of mammalian cell types. However, the role of this acid-sensitive outwardly rectifying (ASOR) anion channel is not known. In human epithelial HeLa cells, reduction in extracellular pH below 5 rapidly activated anion-selective whole-cell currents. The currents exhibited strong outward rectification, activation kinetics at positive potentials, low-field anion selectivity, and sensitivity to 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS) and phloretin. When outside-out patches were exposed to acidic bathing solution, single-channel events of the anion channel could be observed. The unitary conductance was 4.8 pS in the voltage range between -80 and +80 mV. The single-channel activity prominently increased with depolarization and was suppressed by DIDS or phloretin. After 1-h incubation in acidic solution (pH 4.5), a significant population of HeLa cells suffered from necrotic cell injury characterized by stainability with propidium iodide and lack of caspase-3 activation. Upon exposure to acidic solution, HeLa cells exhibited immediate, persistent swelling. Both the necrotic volume increase and cell injury induced by extracellular acidification were inhibited by DIDS or phloretin. Therefore, it is concluded that the ASOR anion channel is involved in the genesis of necrotic cell injury induced by acidosis in human epithelial cells.
Collapse
Affiliation(s)
- Hai-Yan Wang
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | | | | | | |
Collapse
|
36
|
Abbracchio MP, Burnstock G, Boeynaems JM, Barnard EA, Boyer JL, Kennedy C, Knight GE, Fumagalli M, Gachet C, Jacobson KA, Weisman GA. International Union of Pharmacology LVIII: update on the P2Y G protein-coupled nucleotide receptors: from molecular mechanisms and pathophysiology to therapy. Pharmacol Rev 2006; 58:281-341. [PMID: 16968944 PMCID: PMC3471216 DOI: 10.1124/pr.58.3.3] [Citation(s) in RCA: 999] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
There have been many advances in our knowledge about different aspects of P2Y receptor signaling since the last review published by our International Union of Pharmacology subcommittee. More receptor subtypes have been cloned and characterized and most orphan receptors de-orphanized, so that it is now possible to provide a basis for a future subdivision of P2Y receptor subtypes. More is known about the functional elements of the P2Y receptor molecules and the signaling pathways involved, including interactions with ion channels. There have been substantial developments in the design of selective agonists and antagonists to some of the P2Y receptor subtypes. There are new findings about the mechanisms underlying nucleotide release and ectoenzymatic nucleotide breakdown. Interactions between P2Y receptors and receptors to other signaling molecules have been explored as well as P2Y-mediated control of gene transcription. The distribution and roles of P2Y receptor subtypes in many different cell types are better understood and P2Y receptor-related compounds are being explored for therapeutic purposes. These and other advances are discussed in the present review.
Collapse
Affiliation(s)
- Maria P Abbracchio
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Conde SV, Obeso A, Vicario I, Rigual R, Rocher A, Gonzalez C. Caffeine inhibition of rat carotid body chemoreceptors is mediated by A2A and A2B adenosine receptors. J Neurochem 2006; 98:616-28. [PMID: 16805851 DOI: 10.1111/j.1471-4159.2006.03912.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Caffeine, an unspecific antagonist of adenosine receptors, is commonly used to treat the apnea of prematurity. We have defined the effects of caffeine on the carotid body (CB) chemoreceptors, the main peripheral controllers of breathing, and identified the adenosine receptors involved. Caffeine inhibited basal (IC50, 210 microm) and low intensity (PO2 approximately 66 mm Hg/30 mm K+) stimulation-induced release of catecholamines from chemoreceptor cells in intact preparations of rat CB in vitro. Opposite to caffeine, 5'-(N-ethylcarboxamido)adenosine (NECA; an A2 agonist) augmented basal and low-intensity hypoxia-induced release. 2-p-(2-Carboxyethyl)phenethyl-amino-5'-N-ethylcaboxamido-adenosine hydrochloride (CGS21680), 2-hexynyl-NECA (HE-NECA) and SCH58621 (A2A receptors agents) neither affected catecholamine release nor altered the caffeine effects. The 8-cycle-1,3-dipropylxanthine (DPCPX; an A1/A2B antagonist) and 8-(4-{[(4-cyanophenyl)carbamoylmethyl]-oxy}phenyl)-1,3-di(n-propyl)xanthine (MRS1754; an A2B antagonist) mimicking of caffeine indicated that caffeine effects are mediated by A2B receptors. Immunocytochemical A2B receptors were located in tyrosine hydroxylase positive chemoreceptor cells. Caffeine reduced by 52% the chemosensory discharges elicited by hypoxia in the carotid sinus nerve. Inhibition had two components with pharmacological analysis indicating that A2A and A2B receptors mediate, respectively, the low (17 x 10(-9) m) and high (160 x 10(-6) m) IC50 effects. It is concluded that endogenous adenosine, via presynaptic A2B and postsynaptic A2A receptors, can exert excitatory effects on the overall output of the rat CB chemoreceptors.
Collapse
Affiliation(s)
- S V Conde
- Departamento de Bioquímica y Biología Molecular y Fisiología/Instituto de Biología y Genética Molecular, Universidad de Valladolid/Consejo Superior de Investigaciones Científicas, Facultad de Medicina, Valladolid, Spain
| | | | | | | | | | | |
Collapse
|
38
|
Liu HT, Tashmukhamedov BA, Inoue H, Okada Y, Sabirov RZ. Roles of two types of anion channels in glutamate release from mouse astrocytes under ischemic or osmotic stress. Glia 2006; 54:343-57. [PMID: 16883573 DOI: 10.1002/glia.20400] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Astrocytes release glutamate upon hyperexcitation in the normal brain, and in response to pathologic insults such as ischemia and trauma. In our experiments, both hypotonic and ischemic stimuli caused the release of glutamate from cultured mouse astrocytes, which occurred with little or no contribution of gap junction hemichannels, vesicle-mediated exocytosis, or reversed operation of the Na-dependent glutamate transporter. Cell swelling and chemical ischemia activated, in cell-attached membrane patches, anionic channels with large unitary conductance (approximately 400 pS) and inactivation kinetics at potentials more positive than +20 mV or more negative than -20 mV. These properties are different from those of volume-sensitive outwardly rectifying (VSOR) Cl- channels, which were also expressed in these cells and exhibited intermediate unitary conductance (approximately 80 pS) and inactivation kinetics at large positive potentials of more than +40 mV. Both maxi-anion channels and VSOR Cl- channels were permeable to glutamate with permeability ratios of glutamate to chloride of 0.21 +/- 0.07 and 0.15 +/- 0.01, respectively. However, the release of glutamate was significantly more sensitive to Gd3+, a blocker of maxi-anion channels, than to phloretin, a blocker of VSOR Cl- channels. We conclude that these two channels jointly represent a major conductive pathway for the release of glutamate from swollen and ischemia-challenged astrocytes, with the contribution of maxi-anion channels being predominant.
Collapse
Affiliation(s)
- Hong-Tao Liu
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | | | | | | | | |
Collapse
|
39
|
Locovei S, Wang J, Dahl G. Activation of pannexin 1 channels by ATP through P2Y receptors and by cytoplasmic calcium. FEBS Lett 2005; 580:239-44. [PMID: 16364313 DOI: 10.1016/j.febslet.2005.12.004] [Citation(s) in RCA: 422] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2005] [Accepted: 12/01/2005] [Indexed: 11/24/2022]
Abstract
The ability for long-range communication through intercellular calcium waves is inherent to cells of many tissues. A dual propagation mode for these waves includes passage of IP3 through gap junctions as well as an extracellular pathway involving ATP. The wave can be regenerative and include ATP-induced ATP release via an unknown mechanism. Here, we show that pannexin 1 channels can be activated by extracellular ATP acting through purinergic receptors of the P2Y group as well as by cytoplasmic calcium. Based on its properties, including ATP permeability, pannexin 1 may be involved in both initiation and propagation of calcium waves.
Collapse
Affiliation(s)
- Silviu Locovei
- Department of Physiology and Biophysics, University of Miami, School of Medicine, P.O. Box 016430, 1600 NW 10th Avenue, Miami, FL 33136, USA
| | | | | |
Collapse
|
40
|
Sabirov RZ, Okada Y. ATP release via anion channels. Purinergic Signal 2005; 1:311-28. [PMID: 18404516 PMCID: PMC2096548 DOI: 10.1007/s11302-005-1557-0] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Revised: 07/19/2005] [Accepted: 07/26/2005] [Indexed: 11/30/2022] Open
Abstract
ATP serves not only as an energy source for all cell types but as an 'extracellular messenger' for autocrine and paracrine signalling. It is released from the cell via several different purinergic signal efflux pathways. ATP and its Mg(2+) and/or H(+) salts exist in anionic forms at physiological pH and may exit cells via some anion channel if the pore physically permits this. In this review we survey experimental data providing evidence for and against the release of ATP through anion channels. CFTR has long been considered a probable pathway for ATP release in airway epithelium and other types of cells expressing this protein, although non-CFTR ATP currents have also been observed. Volume-sensitive outwardly rectifying (VSOR) chloride channels are found in virtually all cell types and can physically accommodate or even permeate ATP(4-) in certain experimental conditions. However, pharmacological studies are controversial and argue against the actual involvement of the VSOR channel in significant release of ATP. A large-conductance anion channel whose open probability exhibits a bell-shaped voltage dependence is also ubiquitously expressed and represents a putative pathway for ATP release. This channel, called a maxi-anion channel, has a wide nanoscopic pore suitable for nucleotide transport and possesses an ATP-binding site in the middle of the pore lumen to facilitate the passage of the nucleotide. The maxi-anion channel conducts ATP and displays a pharmacological profile similar to that of ATP release in response to osmotic, ischemic, hypoxic and salt stresses. The relation of some other channels and transporters to the regulated release of ATP is also discussed.
Collapse
Affiliation(s)
- Ravshan Z. Sabirov
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585 Japan
| | - Yasunobu Okada
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585 Japan
| |
Collapse
|
41
|
Sabirov RZ, Sheiko T, Liu H, Deng D, Okada Y, Craigen WJ. Genetic demonstration that the plasma membrane maxianion channel and voltage-dependent anion channels are unrelated proteins. J Biol Chem 2005; 281:1897-904. [PMID: 16291750 DOI: 10.1074/jbc.m509482200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The maxianion channel is widely expressed in many cell types, where it fulfills a general physiological function as an ATP-conductive gate for cell-to-cell purinergic signaling. Establishing the molecular identity of this channel is crucial to understanding the mechanisms of regulated ATP release. A mitochondrial porin (voltage-dependent anion channel (VDAC)) located in the plasma membrane has long been considered as the molecule underlying the maxianion channel activity, based upon similarities in the biophysical properties of these two channels and the purported presence of VDAC protein in the plasma membrane. We have deleted each of the three genes encoding the VDAC isoforms individually and collectively and demonstrate that maxianion channel (approximately 400 picosiemens) activity in VDAC-deficient mouse fibroblasts is unaltered. The channel activity is similar in VDAC1/VDAC3-double-deficient cells and in double-deficient cells with the VDAC2 protein depleted by RNA interference. VDAC deletion slightly down-regulated, but never abolished, the swelling-induced ATP release. The lack of correlation between VDAC protein expression and maxianion channel activity strongly argues against the long held hypothesis of plasmalemmal VDAC being the maxianion channel.
Collapse
Affiliation(s)
- Ravshan Z Sabirov
- Department of Cell Physiology, National Institute for Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8585, Japan.
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
In the kidney, macula densa cells communicate with the mesangial cell-afferent arteriolar smooth muscle cell complex through ATP signaling. This signaling process involves release of ATP across the macula densa basolateral membrane through a maxi anion channel and the interaction of ATP with purinergic P2 receptors.
Collapse
Affiliation(s)
- Peter Komlosi
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | | | | |
Collapse
|
43
|
Sabirov RZ, Okada Y. Wide nanoscopic pore of maxi-anion channel suits its function as an ATP-conductive pathway. Biophys J 2005; 87:1672-85. [PMID: 15345546 PMCID: PMC1304572 DOI: 10.1529/biophysj.104.043174] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The newly proposed function of the maxi-anion channel as a conductive pathway for ATP release requires that its pore is sufficiently large to permit passage of a bulky ATP(4-) anion. We found a linear relationship between relative permeability of organic anions of different size and their relative ionic mobility (measured as the ratio of ionic conductance) with a slope close to 1, suggesting that organic anions tested with radii up to 0.49 nm (lactobionate) move inside the channel by free diffusion. In the second approach, we, for the first time, succeeded in pore sizing by the nonelectrolyte exclusion method in single-channel patch-clamp experiments. The cutoff radii of PEG molecules that could access the channel from intracellular (1.16 nm) and extracellular (1.42 nm) sides indicated an asymmetry of the two entrances to the channel pore. Measurements by symmetrical two-sided application of PEG molecules yielded an average functional pore radius of approximately 1.3 nm. These three estimates are considerably larger than the radius of ATP(4-) (0.57-0.65 nm) and MgATP(2-) (approximately 0.60 nm). We therefore conclude that the nanoscopic maxi-anion channel pore provides sufficient room to accommodate ATP and is well suited to its function as a conductive pathway for ATP release in cell-to-cell communication.
Collapse
Affiliation(s)
- Ravshan Z Sabirov
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan.
| | | |
Collapse
|
44
|
Abstract
Cells release adenosine 5'-triphosphate (ATP) into the extracellular space in response to various stimuli. This released ATP plays an important physiological role in cell-to-cell signal transduction. The bulk ATP concentration can be detected using a conventional luciferin-luciferase assay. However, the ATP concentration in the vicinity of the cell surface is often different from the bulk concentration because of its rapid degradation by ecto-ATPases and because of delayed diffusion due to unstirred layer effects. Here, we describe a simple biosensor method to measure the local ATP concentration on the cell surface in real time. The method is based on the ATP-dependent opening of ligand-gated cation channels of purinergic P2X receptors expressed in undifferentiated pheochromocytoma (PC12) cells or in human embryonic kidney 293 (HEK293) cells stably transfected with recombinant P2X2 purinergic receptors. Under the whole-cell configuration of patch-clamp, a sensor PC12 cell or HEK293 is positioned within the proximity of a target cell, and the P2X-mediated currents induced by ATP released from a given site on the target cell surface is measured. The ATP release is quantified by a calibration procedure utilizing local puff applications of ATP at preset concentrations.
Collapse
Affiliation(s)
- Seiji Hayashi
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | | | | | | | | |
Collapse
|
45
|
Ternovsky VI, Okada Y, Sabirov RZ. Sizing the pore of the volume-sensitive anion channel by differential polymer partitioning. FEBS Lett 2004; 576:433-6. [PMID: 15498575 DOI: 10.1016/j.febslet.2004.09.051] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Revised: 09/15/2004] [Accepted: 09/24/2004] [Indexed: 10/26/2022]
Abstract
Partitioning of ethylene glycol and its polymeric forms into the pore of the volume-sensitive outwardly rectifying (VSOR) anion channel was studied to assess the pore size. Polyethylene glycol (PEG) PEG 200-300 (Rh = 0.27-0.53 nm) effectively suppressed the single-channel currents, whereas PEG 400-4000 (Rh = 0.62-1.91 nm) had little or no effect. Since all the molecules tested effectively decreased electric conductivity of the bulk solution, the observed differential effects between PEG 200-300 and PEG 400-4000 on the VSOR single-channel current are due to their limited partitioning into the channel lumen. The cut-off radius of the VSOR channel pore was assessed to be 0.63 nm.
Collapse
Affiliation(s)
- Vadim I Ternovsky
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | | | | |
Collapse
|
46
|
Dutta AK, Sabirov RZ, Uramoto H, Okada Y. Role of ATP-conductive anion channel in ATP release from neonatal rat cardiomyocytes in ischaemic or hypoxic conditions. J Physiol 2004; 559:799-812. [PMID: 15272030 PMCID: PMC1665184 DOI: 10.1113/jphysiol.2004.069245] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
It is known that the level of ATP in the interstitial spaces within the heart during ischaemia or hypoxia is elevated due to its release from a number of cell types, including cardiomyocytes. However, the mechanism by which ATP is released from these myocytes is not known. In this study, we examined a possible involvement of the ATP-conductive maxi-anion channel in ATP release from neonatal rat cardiomyocytes in primary culture upon ischaemic, hypoxic or hypotonic stimulation. Using a luciferin-luciferase assay, it was found that ATP was released into the bulk solution when the cells were subjected to chemical ischaemia, hypoxia or hypotonic stress. The swelling-induced ATP release was inhibited by the carboxylate- and stilbene-derivative anion channel blockers, arachidonic acid and Gd3+, but not by glibenclamide. The local concentration of ATP released near the cell surface of a single cardiomyocyte, measured by a biosensor technique, was found to exceed the micromolar level. Patch-clamp studies showed that ischaemia, hypoxia or hypotonic stimulation induced the activation of single-channel events with a large unitary conductance (approximately 390 pS). The channel was selective to anions and showed significant permeability to ATP4- (PATP/PCl approximately 0.1) and MgATP2- (PATP/PCl approximately 0.16). The channel activity exhibited pharmacological properties essentially identical to those of ATP release. These results indicate that neonatal rat cardiomyocytes respond to ischaemia, hypoxia or hypotonic stimulation with ATP release via maxi-anion channels.
Collapse
Affiliation(s)
- Amal K Dutta
- Department of Cell Physiology, National Institute for Physiological Sciences, Myodaiji-cho, Okazaki 444-8585, Japan
| | | | | | | |
Collapse
|