1
|
Liang WZ, Hsieh KW, Yang ZD, Sun GC. Induction of Ca 2+ signaling and cytotoxic responses of human lung fibroblasts upon an antihistamine drug oxatomide treatment and evaluating the protective effects of Ca 2+ chelating. Fundam Clin Pharmacol 2025; 39:e13040. [PMID: 39431647 DOI: 10.1111/fcp.13040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 09/05/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Oxatomide, an antihistamine drug of the diphenylmethylpiperazine family, has anti-inflammatory effects in airway disease. Because oxatomide was shown to cause diverse physiological responses in several cell models, the impact of oxatomide on Ca2+ signaling and its related physiological effects has not been explored in IMR-90 human fetal lung fibroblasts. OBJECTIVES This study assessed the effect of oxatomide on cell viability and intracellular free Ca2+ concentrations ([Ca2+]i) and examined whether oxatomide-induced cytotoxicity through Ca2+ signaling in IMR-90 cells. METHODS Cell viability was measured by the cell proliferation reagent (WST-1). [Ca2+]i was measured by the Ca2+-sensitive fluorescent dye fura-2. RESULTS Oxatomide (10-40 μM) concentration dependently reduced cell viability and induced [Ca2+]i rises in IMR-90 cells. This cytotoxic effect was reversed by chelation of cytosolic Ca2+ with BAPTA-AM. In terms of Ca2+ signaling, oxatomide-caused Ca2+ entry was inhibited by modulators of store-operated Ca2+ channels (2-APB and SKF96365) and protein kinase C (PKC) inhibitor (GF109203X). Furthermore, oxatomide-induced Ca2+ influx was confirmed by Mn2+-induced quench of fura-2 fluorescence. In a Ca2+-free medium, preincubation with the endoplasmic reticulum Ca2+ pump inhibitor thapsigargin inhibited oxatomide-evoked [Ca2+]i rises. Conversely, treatment with oxatomide abolished thapsigargin-induced [Ca2+]i rises. Inhibition of phospholipase C (PLC) with U73122 also inhibited oxatomide-caused [Ca2+]i rises. CONCLUSION In IMR-90 cells, oxatomide-induced cytotoxicity by preceding [Ca2+]i rises involving PKC-sensitive store-operated Ca2+ entry and PLC-dependent Ca2+ release from the endoplasmic reticulum. BAPTA-AM, with its Ca2+ chelating effects, may be a potential compound for preventing oxatomide-induced cytotoxicity.
Collapse
Affiliation(s)
- Wei-Zhe Liang
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Yanpu Township, Pingtung County, Taiwan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Kai-Wei Hsieh
- Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Zong-Da Yang
- Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Gwo-Ching Sun
- Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
2
|
Goriounova AS, Flori Sassano M, Wrennall JA, Tarran R. ELD607 specifically traffics Orai1 to the lysosome leading to inhibition of store operated calcium entry. Cell Calcium 2024; 123:102945. [PMID: 39191091 DOI: 10.1016/j.ceca.2024.102945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
Orai1 is a plasma membrane Ca2+ channel involved in store operated calcium entry (SOCE). SOCE can regulate cell growth, exocytosis, gene expression and inflammation. We previously found that short palate lung and nasal epithelial clone 1's (SPLUNC1) sixth α-helix (α6) bound Orai1 to inhibit SOCE. SPLUNC1 was not proteolytically stable, so we developed ELD607, an 11 amino acid peptide based on SPLUNC1's α6 region which was more stable and more potent than SPLUNC1/α6. Here, we studied ELD607's mechanism of action. We overexpressed either Orai1-HA or Orai1-YFP in HEK293T cells to probe ELD607-Orai1 interactions by confocal microscopy. We also measured changes in Fluo-4 fluorescence in a multiplate reader as a marker of cytoplasmic Ca2+ levels. ELD607 internalized Orai1 independently of STIM1. Both 15 min and 3 h exposure to ELD607 similarly depleted Orai1 in the plasma membrane. However, 3 h exposure to ELD607 yielded greater inhibition of SOCE. ELD607 continued to colocalize with Orai1 after internalization and this process was dependent on the presence of the ubiquitin ligase NEDD4.2. Similarly, ELD607 increased the colocalization between Orai1 and ubiquitin. ELD607 also increased the colocalization between Orai1 and Rab5 and 7, but not Rab11, suggesting that Orai1 trafficked through early and late but not recycling endosomes. Finally, ELD607 caused Orai1, but not Orai2, Orai3, or STIM1 to traffic to lysosomes. We conclude that ELD607 rapidly binds to Orai1 and works in an identical fashion as full length SPLUNC1 by internalizing Orai1 and sending it to lysosomes, leading to a decrease in SOCE.
Collapse
Affiliation(s)
- Alexandra S Goriounova
- Department of Pharmacology, The University of North Carolina at Chapel Hill, NC 27599, USA
| | - M Flori Sassano
- Division of Genetic, Environmental and Inhalational Disease, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA
| | - Joe A Wrennall
- Department of Cell Biology & Physiology, The University of North Carolina at Chapel Hill, NC 27599, USA
| | - Robert Tarran
- Division of Genetic, Environmental and Inhalational Disease, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA.
| |
Collapse
|
3
|
Goriounova AS, Gilmore RC, Wrennall JA, Tarran R. Super resolution microscopy analysis reveals increased Orai1 activity in asthma and cystic fibrosis lungs. J Cyst Fibros 2023; 22:161-171. [PMID: 35961837 PMCID: PMC9982747 DOI: 10.1016/j.jcf.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/28/2022]
Abstract
QUESTION In diseases such as asthma and cystic fibrosis (CF), the immune response is dysregulated and the lung is chronically inflamed. Orai1 activation is required for the initiation and persistence of inflammation. However, Orai1 expression in the lung is poorly understood. We therefore tested the hypothesis that Orai1 expression was upregulated in asthmatic and CF lungs. MATERIALS AND METHODS We used LungMAP to analyze single-cell RNAseq data of Orai1 and stromal interaction molecule 1 (STIM1) expression in normal human lungs. We then performed RNAscope analysis and immunostaining on lung sections from normal, asthma, and CF donors. We imaged sections by confocal and super resolution microscopy, and analyzed Orai1 and STIM1 expression in different pulmonary cell types. RESULTS Orai1 was broadly-expressed, but expression was greatest in immune cells. At mRNA and protein levels, there were no consistent trends in expression levels between the three phenotypes. Orai1 must interact with STIM1 in order to activate and conduct Ca2+. We therefore used STIM1/Orai1 co-localization as a marker of Orai1 activity. Using this approach, we found significantly increased co-localization between these proteins in epithelia, interstitial and luminal immune cells, but not alveoli, from asthma and CF lungs. Orai1 also aggregates as part of its activation process. Using super resolution microscopy, we also found significantly increased Orai1 aggregation in immune cells from asthmatic and CF lungs. CONCLUSION We found evidence that Orai1 was more active in asthma and CF than normal lungs. These data suggest that Orai1 is a relevant target for reducing pulmonary inflammation.
Collapse
Affiliation(s)
| | | | - Joe A Wrennall
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Robert Tarran
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, NC, 27599, USA.
| |
Collapse
|
4
|
Wrennall JA, Ahmad S, Worthington EN, Wu T, Goriounova AS, Voeller AS, Stewart IE, Ghosh A, Krajewski K, Tilley SL, Hickey AJ, Sassano MF, Tarran R. A SPLUNC1 Peptidomimetic Inhibits Orai1 and Reduces Inflammation in a Murine Allergic Asthma Model. Am J Respir Cell Mol Biol 2022; 66:271-282. [PMID: 34807800 PMCID: PMC8937239 DOI: 10.1165/rcmb.2020-0452oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/22/2021] [Indexed: 11/24/2022] Open
Abstract
Orai1 is a plasma membrane Ca2+ channel that mediates store-operated Ca2+ entry (SOCE) and regulates inflammation. Short palate lung and nasal epithelial clone 1 (SPLUNC1) is an asthma gene modifier that inhibits Orai1 and SOCE via its C-terminal α6 region. SPLUNC1 levels are diminished in asthma patient airways. Thus, we hypothesized that inhaled α6 peptidomimetics could inhibit Orai1 and reduce airway inflammation in a murine asthma model. To evaluate α6-Orai1 interactions, we used fluorescent assays to measure Ca2+ signaling, Förster resonance energy transfer, fluorescent recovery after photobleaching, immunostaining, total internal reflection microscopy, and Western blotting. To test whether α6 peptidomimetics inhibited SOCE and decreased inflammation in vivo, wild-type and SPLUNC1-/- mice were exposed to house dust mite (HDM) extract with or without α6 peptide. We also performed nebulization, jet milling, and scanning electron microscopy to evaluate α6 for inhalation. SPLUNC1-/- mice had an exaggerated response to HDM. In BAL-derived immune cells, Orai1 levels increased after HDM exposure in SPLUNC1-/- but not wild-type mice. Inhaled α6 reduced Orai1 levels in mice regardless of genotype. In HDM-exposed mice, α6 dose-dependently reduced eosinophilia and neutrophilia. In vitro, α6 inhibited SOCE in multiple immune cell types, and α6 could be nebulized or jet milled without loss of function. These data suggest that α6 peptidomimetics may be a novel, effective antiinflammatory therapy for patients with asthma.
Collapse
Affiliation(s)
| | | | | | - Tongde Wu
- Department of Cell Biology and Physiology
| | | | | | - Ian E. Stewart
- Center for Engineered Systems, Research Triangle Institute International, Research Triangle Park, North Carolina
| | | | | | - Steven L. Tilley
- Division of Pulmonology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Anthony J. Hickey
- Center for Engineered Systems, Research Triangle Institute International, Research Triangle Park, North Carolina
| | | | | |
Collapse
|
5
|
Kountz TS, Jairaman A, Kountz CD, Stauderman KA, Schleimer RP, Prakriya M. Differential Regulation of ATP- and UTP-Evoked Prostaglandin E 2 and IL-6 Production from Human Airway Epithelial Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:1275-1287. [PMID: 34389624 PMCID: PMC8816324 DOI: 10.4049/jimmunol.2100127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/07/2021] [Indexed: 11/19/2022]
Abstract
The airway epithelial cells (AECs) lining the conducting passageways of the lung secrete a variety of immunomodulatory factors. Among these, PGE2 limits lung inflammation and promotes bronchodilation. By contrast, IL-6 drives intense airway inflammation, remodeling, and fibrosis. The signaling that differentiates the production of these opposing mediators is not understood. In this study, we find that the production of PGE2 and IL-6 following stimulation of human AECs by the damage-associated molecular pattern extracellular ATP shares a common requirement for Ca2+ release-activated Ca2+ (CRAC) channels. ATP-mediated synthesis of PGE2 required activation of metabotropic P2Y2 receptors and CRAC channel-mediated cytosolic phospholipase A2 signaling. By contrast, ATP-evoked synthesis of IL-6 occurred via activation of ionotropic P2X receptors and CRAC channel-mediated calcineurin/NFAT signaling. In contrast to ATP, which elicited the production of both PGE2 and IL-6, the uridine nucleotide, UTP, stimulated PGE2 but not IL-6 production. These results reveal that human AECs employ unique receptor-specific signaling mechanisms with CRAC channels as a signaling nexus to regulate release of opposing immunomodulatory mediators. Collectively, our results identify P2Y2 receptors, CRAC channels, and P2X receptors as potential intervention targets for airway diseases.
Collapse
Affiliation(s)
- Timothy S Kountz
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Amit Jairaman
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Candace D Kountz
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Robert P Schleimer
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL;
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
6
|
De Ita M, Vargas MH, Carbajal V, Ortiz-Quintero B, López-López C, Miranda-Morales M, Barajas-López C, Montaño LM. ATP releases ATP or other nucleotides from human peripheral blood leukocytes through purinergic P2 receptors. Life Sci 2015; 145:85-92. [PMID: 26679107 DOI: 10.1016/j.lfs.2015.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 11/07/2015] [Accepted: 12/05/2015] [Indexed: 12/29/2022]
Abstract
AIMS Almost every eukaryotic cell releases ATP under certain conditions. The idea that ATP induces the release of ATP has been scantly investigated. METHODS We explored this possibility by assessing the rate of exogenous ATP breakdown (measured by phosphates production) by human peripheral blood leukocytes. The role of P2Y and P2X receptors was evaluated pharmacologically, by patch clamp, or by flow cytometry. KEY FINDINGS In mononuclear and/or polymorphonuclear cells, ATP increased phosphates formation in a time- and concentration-dependent manner. Uncoupling of P2Y receptors with N-ethylmaleimide and antagonism of P2Y and P2X receptors through suramin reduced phosphate formation after 500μM ATP, suggesting that part of the phosphate production was due to activation of P2 receptors, with subsequent release of ATP or other nucleotides. Similar results were obtained with UTP and ATPγS. Gadolinium (connexins inhibitor) also significantly reduced the ATP-induced phosphate production. Blockade of P2X receptors with SKF 96365 or NF023 did not modify the phosphate production. In monocytes, 500μM ATP induced inward currents suggestive of P2X1 activation, but higher concentrations (1-5mM) induced inward currents suggestive of P2X7 activation. We discarded a role of adenosine in the ATP-evoked nucleotides release. Flow cytometry identified that almost all mononuclear and polymorphonuclear cells expressed P2Y1,2,4,6,11 receptors. SIGNIFICANCE 500μM ATP induced the release of ATP or other nucleotides through activation of P2Y2,4,6,11 receptors in human leukocytes, and probably via P2X receptors at higher concentrations. This ATP-induced nucleotides release constitutes a potential mechanism leading to amplification of ATP signaling.
Collapse
Affiliation(s)
- Marlon De Ita
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, México, DF, México
| | - Mario H Vargas
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias, México, DF, México
| | - Verónica Carbajal
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias, México, DF, México
| | - Blanca Ortiz-Quintero
- Departamento de Investigación en Bioquímica, Instituto Nacional de Enfermedades Respiratorias, México, DF, México
| | - Cintya López-López
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosí, SLP, México
| | - Marcela Miranda-Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Carlos Barajas-López
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosí, SLP, México
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, México, DF, México.
| |
Collapse
|
7
|
Chávez J, Vargas MH, Rebollar-Ayala DC, Díaz-Hernández V, Cruz-Valderrama JE, Flores-Soto E, Flores-García M, Jiménez-Vargas NN, Barajas-López C, Montaño LM. Inhibition of extracellular nucleotides hydrolysis intensifies the allergic bronchospasm. A novel protective role of ectonucleotidases. Allergy 2013; 68:462-71. [PMID: 23414231 DOI: 10.1111/all.12113] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2012] [Indexed: 01/31/2023]
Abstract
BACKGROUND Nucleotides released to the extracellular space stimulate purinergic receptors, and their effects are modulated by ectonucleotidases. The role of ATP in the allergic bronchospasm has been scantly studied. METHODS We used several techniques (plethysmography, organ baths, confocal microscopy, RT-PCR, ATP measurement) to explore the role of nucleotides and ectonucleotidases in the allergic bronchospasm in guinea pigs. RESULTS While allergenic challenge with a low-dose ovalbumin (OVA) only produced a small bronchospasm (~2-fold the basal lung resistance), previous inhibition of ectonucleotidases by ARL-67156 greatly intensified this response (~11-fold the basal lung resistance, with 44% mortality). Bronchoalveolar lavage fluid obtained during this bronchospasm contained increased ATP concentration. This potentiation was abolished by antagonism of purinergic receptors (suramin+RB2) or TXA2 receptor (SQ29548), or by intratracheal apyrase. In tracheal rings and lung parenchyma strips, OVA caused a concentration-dependent contraction. Suramin+RB2 or levamisole produced a significant rightward displacement of this response, and ARL-67156 did not modify it. Platelets stimulated with OVA released ATP. Confocal images of nonsensitized tracheas showed slight fluorescence for P2Y6 receptors in epithelium and none for P2Y4 . Sensitized animals showed strong fluorescence to both receptors and to alkaline phosphatase in the airway epithelium. This correlated with a large increment in mRNA for P2Y4 and P2Y6 receptors in sensitized animals. CONCLUSIONS Nucleotides greatly potentiate the allergic bronchospasm when ectonucleotidases activity is diminished, and this effect is probably favored by the upregulation of P2Y4 and P2Y6 receptors in airway epithelium during sensitization. These results prompt for further research on these mechanisms in human asthma.
Collapse
Affiliation(s)
- J. Chávez
- Departamento de Investigación en Hiperreactividad Bronquial; Instituto Nacional de Enfermedades Respiratorias; México; DF; México
| | - M. H. Vargas
- Departamento de Investigación en Hiperreactividad Bronquial; Instituto Nacional de Enfermedades Respiratorias; México; DF; México
| | - D. C. Rebollar-Ayala
- Departamento de Farmacología; Facultad de Medicina; Universidad Nacional Autónoma de México; México; DF; México
| | - V. Díaz-Hernández
- Departamento de Embriología; Facultad de Medicina; Universidad Nacional Autónoma de México; México; DF; México
| | - J. E. Cruz-Valderrama
- Departamento de Farmacología; Facultad de Medicina; Universidad Nacional Autónoma de México; México; DF; México
| | - E. Flores-Soto
- Departamento de Farmacología; Facultad de Medicina; Universidad Nacional Autónoma de México; México; DF; México
| | - M. Flores-García
- Departamento de Biología Molecular; Instituto Nacional de Cardiología; México; DF; México
| | - N. N. Jiménez-Vargas
- División de Biología Molecular; Instituto Potosino de Investigación Científica y Tecnológica; San Luis Potosí; S.L.P.; México
| | - C. Barajas-López
- División de Biología Molecular; Instituto Potosino de Investigación Científica y Tecnológica; San Luis Potosí; S.L.P.; México
| | - L. M. Montaño
- Departamento de Farmacología; Facultad de Medicina; Universidad Nacional Autónoma de México; México; DF; México
| |
Collapse
|
8
|
Erb L, Weisman GA. Coupling of P2Y receptors to G proteins and other signaling pathways. ACTA ACUST UNITED AC 2012; 1:789-803. [PMID: 25774333 DOI: 10.1002/wmts.62] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
P2Y receptors are G protein-coupled receptors (GPCRs) that are activated by adenine and uridine nucleotides and nucleotide sugars. There are eight subtypes of P2Y receptors (P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, and P2Y14), which activate intracellular signaling cascades to regulate a variety of cellular processes, including proliferation, differentiation, phagocytosis, secretion, nociception, cell adhesion, and cell migration. These signaling cascades operate mainly by the sequential activation or deactivation of heterotrimeric and monomeric G proteins, phospholipases, adenylyl and guanylyl cyclases, protein kinases, and phosphodiesterases. In addition, there are numerous ion channels, cell adhesion molecules, and receptor tyrosine kinases that are modulated by P2Y receptors and operate to transmit an extracellular signal to an intracellular response.
Collapse
Affiliation(s)
- Laurie Erb
- Department of Biochemistry, Life Sciences Center, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Gary A Weisman
- Department of Biochemistry, Life Sciences Center, University of Missouri-Columbia, Columbia, MO 65211, USA
| |
Collapse
|
9
|
Palmer ML, Peitzman ER, Maniak PJ, Sieck GC, Prakash YS, O'Grady SM. K(Ca)3.1 channels facilitate K+ secretion or Na+ absorption depending on apical or basolateral P2Y receptor stimulation. J Physiol 2011; 589:3483-94. [PMID: 21606112 DOI: 10.1113/jphysiol.2011.207548] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human mammary epithelial (HME) cells express several P2Y receptor subtypes located in both apical and basolateral membranes. Apical UTP or ATP-γ-S stimulation of monolayers mounted in Ussing chambers evoked a rapid, but transient decrease in short circuit current (I(sc)), consistent with activation of an apical K+ conductance. In contrast, basolateral P2Y receptor stimulation activated basolateral K+ channels and increased transepithelial Na+ absorption. Chelating intracellular Ca2+ using the membrane-permeable compound BAPTA-AM, abolished the effects of purinoceptor activation on I(sc). Apical pretreatment with charybdotoxin also blocked the I(sc) decrease by >90% and similar magnitudes of inhibition were observed with clotrimazole and TRAM-34. In contrast, iberiotoxin and apamin did not block the effects of apical P2Y receptor stimulation. Silencing the expression of K(Ca)3.1 produced ∼70% inhibition of mRNA expression and a similar reduction in the effects of apical purinoceptor agonists on I(sc). In addition, silencing P2Y2 receptors reduced the level of P2Y2 mRNA by 75% and blocked the effects of ATP-γ-S by 65%. These results suggest that P2Y2 receptors mediate the effects of purinoceptor agonists on K+ secretion by regulating the activity of K(Ca)3.1 channels expressed in the apical membrane of HME cells. The results also indicate that release of ATP or UTP across the apical or basolateral membrane elicits qualitatively different effects on ion transport that may ultimately determine the [Na+]/[K+] composition of fluid within the mammary ductal network.
Collapse
Affiliation(s)
- Melissa L Palmer
- Biology Program, College of Biological Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Airway mucus plugging has long been recognized as a principal cause of death in asthma. However, molecular mechanisms of mucin overproduction and secretion have not been understood until recently. These mechanisms are reviewed together with ongoing investigations relating them to lung pathophysiology. RECENT FINDINGS Of the five secreted gel-forming mucins in mammals, only MUC5AC and MUC5B are produced in significant quantities in intrapulmonary airways. MUC5B is the principal gel-forming mucin at baseline in small airways of humans and mice, and therefore likely performs most homeostatic clearance functions. MUC5AC is the principal gel-forming mucin upregulated in airway inflammation and is under negative control by forkhead box a2 (Foxa2) and positive control by hypoxia inducible factor-1 (HIF-1). Mucin secretion is regulated separately from production, principally by extracellular triphosphate nucleotides that bind P2Y2 receptors on the lumenal surface of airway secretory cells, generating intracellular second messengers that activate the exocytic proteins, Munc13-2 and synaptotagmin-2. SUMMARY Markedly upregulated production of MUC5AC together with stimulated secretion leads to airflow obstruction in asthma. As MUC5B appears to mediate homeostatic functions, it may be possible to selectively inhibit MUC5AC production without impairing airway function. The precise roles of mucin hypersecretion in asthma symptoms such as dyspnea and cough and in physiologic phenomena such as airway hyperresponsiveness remain to be defined.
Collapse
|
11
|
Tuvim MJ, Mospan AR, Burns KA, Chua M, Mohler PJ, Melicoff E, Adachi R, Ammar-Aouchiche Z, Davis CW, Dickey BF. Synaptotagmin 2 couples mucin granule exocytosis to Ca2+ signaling from endoplasmic reticulum. J Biol Chem 2009; 284:9781-7. [PMID: 19208631 DOI: 10.1074/jbc.m807849200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Synaptotagmin 2 (Syt2) functions as a low affinity, fast exocytic Ca(2+) sensor in neurons, where it is activated by Ca(2+) influx through voltage-gated channels. Targeted insertion of lacZ into the mouse syt2 locus reveals expression in mucin-secreting goblet cells of the airways. In these cells, rapid Ca(2+) entry from the extracellular medium does not contribute significantly to stimulated secretion (Davis, C. W., and Dickey, B. F. (2008) Annu. Rev. Physiol. 70, 487-512). Nonetheless, Syt2(-/-) mice show a severe defect in acute agonist-stimulated airway mucin secretion, and Syt2(+/-) mice show a partial defect. In contrast to Munc13-2(-/-) mice (Zhu, Y., Ehre, C., Abdullah, L. H., Sheehan, J. K., Roy, M., Evans, C. M., Dickey, B. F., and Davis, C. W. (2008) J. Physiol. (Lond.) 586, 1977-1992), Syt2(-/-) mice show no spontaneous mucin accumulation, consistent with the inhibitory action of Syt2 at resting cytoplasmic Ca(2+) in neurons. In human airway goblet cells, inositol trisphosphate receptors are found in rough endoplasmic reticulum that closely invests apical mucin granules, consistent with the known dependence of exocytic Ca(2+) signaling on intracellular stores in these cells. Hence, Syt2 can serve as an exocytic sensor for diverse Ca(2+) signaling systems, and its levels are limiting for stimulated secretory function in airway goblet cells.
Collapse
Affiliation(s)
- Michael J Tuvim
- Department of Pulmonary Medicine, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Dong X, Smoll EJ, Ko KH, Lee J, Chow JY, Kim HD, Insel PA, Dong H. P2Y receptors mediate Ca2+ signaling in duodenocytes and contribute to duodenal mucosal bicarbonate secretion. Am J Physiol Gastrointest Liver Physiol 2009; 296:G424-32. [PMID: 19074643 PMCID: PMC2643905 DOI: 10.1152/ajpgi.90314.2008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Since little is known about the role of P2Y receptors (purinoceptors) in duodenal mucosal bicarbonate secretion (DMBS), we sought to investigate the expression and function of these receptors in duodenal epithelium. Expression of P2Y(2) receptors was detected by RT-PCR in mouse duodenal epithelium and SCBN cells, a duodenal epithelial cell line. UTP, a P2Y(2)-receptor agonist, but not ADP (10 microM), significantly induced murine duodenal short-circuit current and DMBS in vitro; these responses were abolished by suramin (300 microM), a P2Y-receptor antagonist, or 2-aminoethoxydiphenyl borate (2-APB; 100 microM), a store-operated channel blocker. Mucosal or serosal addition of UTP induced a comparable DMBS in wild-type mice, but markedly impaired response occurred in P2Y(2) knockout mice. Acid-stimulated DMBS in vivo was significantly inhibited by suramin (1 mM) or PPADS (30 microM). Both ATP and UTP, but not ADP (1 microM), raised cytoplasmic-free Ca(2+) concentrations ([Ca(2+)](cyt)) with similar potencies in SCBN cells. ATP-induced [Ca(2+)](cyt) was attenuated by U-73122 (10 microM), La(3+) (30 microM), or 2-APB (10 microM), but was not significantly affected by nifedipine (10 microM). UTP (1 microM) induced a [Ca(2+)](cyt) transient in Ca(2+)-free solutions, and restoration of external Ca(2+) (2 mM) raised [Ca(2+)](cyt) due to capacitative Ca(2+) entry. La(3+) (30 microM), SK&F96365 (30 microM), and 2-APB (10 microM) inhibited UTP-induced Ca(2+) entry by 92, 87, and 94%, respectively. Taken together, our results imply that activation of P2Y(2) receptors enhances DMBS via elevation of [Ca(2+)](cyt) that likely results from an initial increase in intracellular Ca(2+) release followed by extracellular Ca(2+) entry via store-operated channel.
Collapse
Affiliation(s)
- Xiao Dong
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California
| | - Eric James Smoll
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California
| | - Kwang Hyun Ko
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California
| | - Jonathan Lee
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California
| | - Jimmy Yip Chow
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California
| | - Ho Dong Kim
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California
| | - Paul A. Insel
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California
| | - Hui Dong
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California
| |
Collapse
|
13
|
Jeulin C, Seltzer V, Bailbé D, Andreau K, Marano F. EGF mediates calcium-activated chloride channel activation in the human bronchial epithelial cell line 16HBE14o-: involvement of tyrosine kinase p60c-src. Am J Physiol Lung Cell Mol Physiol 2008; 295:L489-96. [PMID: 18586953 DOI: 10.1152/ajplung.90282.2008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Particulate atmospheric pollutants interact with the human airway epithelium, which releases cytokines, chemokines, and EGF receptor (EGFR) ligands leading to proinflammatory responses. There is little information concerning the short-term effects of EGFR activation by extracellular ligands on ionic regulation of airway surface lining fluids. We identified in the membrane of human epithelial bronchial cells (16HBE14o(-) line) an endogenous calcium- and voltage-dependent, outwardly rectifying small-conductance chloride channel (CACC), and we examined the effects of EGF on CACC activity. Ion channel currents were recorded with the patch-clamp technique. In cell-attached membrane patches, CACC were activated by exposure of the external surface of the cells to physiological concentrations of EGF without any change in cytosolic Ca(2+) concentration ([Ca(2+)](i)) and inhibited by tyrphostin AG-1478 (an inhibitor of EGFR that also blocks EGF-dependent Src family kinase activation). EGF activation of c-Src protein in 16HBE14o(-) cells was observed, and the signaling pathway elicited by EGFR was blocked by tyrphostin AG-1478. In excised inside-out membrane patches CACC were activated by exposure of the cytoplasmic face of the channels to the human recombinant Src(p60(c-src)) kinase with endogenous or exogenous ATP and inhibited by lambda-protein phosphatase. Secretion of EGFR ligands by epithelial airway cells exposed to pollutants would then elicit a rapid and direct ionic response of CACC mediated by EGFR activation via a Src kinase family-dependent signaling pathway.
Collapse
Affiliation(s)
- Claudette Jeulin
- Laboratoire de Cytophysiologie et Toxicologie Cellulaire, case courrier 7073, 3ème étage, T53-54, Université Paris 7 Denis Diderot, 2 Place Jussieu, 75251 Paris Cedex 05, France.
| | | | | | | | | |
Collapse
|
14
|
Yamazaki D, Aoyama M, Ohya S, Muraki K, Asai K, Imaizumi Y. Novel functions of small conductance Ca2+-activated K+ channel in enhanced cell proliferation by ATP in brain endothelial cells. J Biol Chem 2006; 281:38430-9. [PMID: 17062575 DOI: 10.1074/jbc.m603917200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Brain capillary endothelial cells (BCECs) form the blood-brain barrier (BBB), which is essential for maintaining homeostasis of the brain. Net cellular turnover, which results from the balance between cell death and proliferation, is important in maintaining BBB homeostasis. Here we report a novel mechanism that underlies ATP-induced cell proliferation in t-BBEC 117, a cell line derived from bovine brain endothelial cells. Application of 0.1-30 mum ATP to t-BBEC 117 concentration-dependently increased intracellular Ca(2+) concentration ([Ca(2+)](i)) in two phases: an initial transient phase and a later and smaller sustained one. These two phases of [Ca(2+)](i) rise were mainly due to Ca(2+) release and sustained Ca(2+) influx, respectively. The pretreatment with apamin, a selective blocker of small conductance Ca(2+)-activated K(+) channels (SK), significantly reduced both the [Ca(2+)](i) increase and K(+) current induced by ATP. Transcripts corresponding to P2Yx, SK2, and transient receptor potential channels were detected in t-BBEC 117. Knock down of SK2 protein, which was the predominant Ca(2+)-activated K(+) channel expressed in t-BBEC 117, by siRNA significantly reduced both the sustained phase of the [Ca(2+)](i) rise and the K(+) current induced by ATP. Cell proliferation was increased significantly by the presence of the stable ATP analogue ATPgammaS. This effect was blunted by UCL1684, a synthesized SK blocker. In conclusion, in brain endothelial cells ATP-induced [Ca(2+)](i) rise activates SK2 current, and the subsequent membrane hyperpolarization enhances Ca(2+) entry presumably through transient receptor potential channels. This positive feedback mechanism can account for the augmented cell proliferation by ATP.
Collapse
Affiliation(s)
- Daiju Yamazaki
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori, Mizuho-ku, Nagoya 467-8603
| | | | | | | | | | | |
Collapse
|
15
|
Sharma A, Tao X, Gopal A, Ligon B, Steer ML, Perides G. Calcium dependence of proteinase-activated receptor 2 and cholecystokinin-mediated amylase secretion from pancreatic acini. Am J Physiol Gastrointest Liver Physiol 2005; 289:G686-95. [PMID: 15976386 DOI: 10.1152/ajpgi.00342.2004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pancreatic acini secrete digestive enzymes in response to a variety of secretagogues including CCK and agonists acting via proteinase-activated receptor-2 (PAR2). We employed the CCK analog caerulein and the PAR2-activating peptide SLIGRL-NH(2) to compare and contrast Ca(2+) changes and amylase secretion triggered by CCK receptor and PAR2 stimulation. We found that secretion stimulated by both agonists is dependent on a rise in cytoplasmic Ca(2+) concentration ([Ca(2+)](i)) and that this rise in [Ca(2+)](i) reflects both the release of Ca(2+) from intracellular stores and accelerated Ca(2+) influx. Both agonists, at low concentrations, elicit oscillatory [Ca(2+)](i) changes, and both trigger a peak plateau [Ca(2+)](i) change at high concentrations. Although the two agonists elicit similar rates of amylase secretion, the rise in [Ca(2+)](i) elicited by caerulein is greater than that elicited by SLIGRL-NH(2). In Ca(2+)-free medium, the rise in [Ca(2+)](i) elicited by SLIGRL-NH(2) is prevented by the prior addition of a supramaximally stimulating concentration of caerulein, but the reverse is not true; the rise elicited by caerulein is neither prevented nor reduced by prior addition of SLIGRL-NH(2). Both the oscillatory and the peak plateau [Ca(2+)](i) changes that follow PAR2 stimulation are prevented by the phospholipase C (PLC) inhibitor U73122, but U73122 prevents only the oscillatory [Ca(2+)](i) changes triggered by caerulein. We conclude that 1) both PAR2 and CCK stimulation trigger amylase secretion that is dependent on a rise in [Ca(2+)](i) and that [Ca(2+)](i) rise reflects release of calcium from intracellular stores as well as accelerated influx of extracellular calcium; 2) PLC mediates both the oscillatory and the peak plateau rise in [Ca(2+)](i) elicited by PAR2 but only the oscillatory rise in [Ca(2+)](i) elicited by CCK stimulation; and 3) the rate of amylase secretion elicited by agonists acting via different types of receptors may not correlate with the magnitude of the [Ca(2+)](i) rise triggered by those different types of secretagogue.
Collapse
Affiliation(s)
- Anupriya Sharma
- Dept. of Surgery, Tufts-New England Medical Center, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
16
|
Hu CM, Cheng YW, Liao JW, Cheng HW, Kang JJ. Induction of contracture and extracellular Ca2+ influx in cardiac muscle by sanguinarine: a study on cardiotoxicity of sanguinarine. J Biomed Sci 2005; 12:399-407. [PMID: 15920678 DOI: 10.1007/s11373-005-3007-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2004] [Accepted: 02/05/2005] [Indexed: 12/14/2022] Open
Abstract
In this study, the toxic effect of sanguinarine (SANG) on heart was studied with isolated cardiac muscle strip isolated from Wistar rat. SANG induced positive inotropic action followed by contracture on the left ventricle and both atria strips. In addition, SANG dose-dependently inhibited spontaneous beat of the right atrium. SANG-induced contracture was completely suppressed by pretreatment with La3+ or in a Ca2+ free Tyrode solution containing 2.5 mM EGTA. Incubating isolated cardiomyocytes with SANG enhanced the 45Ca2+ influx, which could be inhibited by pretreatment with La3+. However, the SANG-induced 45Ca2+ influx could not be inhibited by pretreatment with other Ca2+ channel blockers, such as nifedipine, verapamil, diltiazem, nickel and manganese, and amiloride. Although antioxidants can inhibit the SANG-induced lipid peroxidation, they could not prevent the SANG-induced contracture. N-acetylcysteine and dithiothreitol, the sulfhydryl reducing agents, were shown to be effective in preventing the SANG-induced contracture. These data suggested that the SANG-induced contracture is caused by the influx of extracellular Ca2+ through a La3+-sensitive Ca2+ channel.
Collapse
Affiliation(s)
- Chien Ming Hu
- Institute of Pharmaceutical Sciences, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|