1
|
Zhao Y, Deng Z, Ma Z, Zhang M, Wang H, Tuo B, Li T, Liu X. Expression alteration and dysfunction of ion channels/transporters in the parietal cells induces gastric diffused mucosal injury. Biomed Pharmacother 2022; 148:112660. [PMID: 35276516 DOI: 10.1016/j.biopha.2022.112660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 11/26/2022] Open
Abstract
Gastric mucosal injuries include focal and diffused injuries, which do and do not change the cell differentiation pattern. Parietal cells loss is related to the occurrence of gastric mucosal diffused injury, with two phenotypes of spasmolytic polypeptide-expressing metaplasia and neuroendocrine cell hyperplasia, which is the basis of gastric cancer and gastric neuroendocrine tumor respectively. Multiple ion channels and transporters are located and expressed in the parietal cells, which is not only regulate the gastric acid-base homeostasis, but also regulate the growth and development of parietal cells. Therefore, alteration and dysregulation of ion channels and transporters in the parietal cells impairs the morphology and physiological functions of stomach, resulted in gastric diffused mucosal damage. In this review, multiple ion channels and transporters in parietal cells, including K+ channels, aquaporins, Cl- channels, Na+/H+ transporters, and Cl-/HCO3- transporters are described, and their roles in gastric diffused mucosal injury are discussed. We hope to drive researcher's attention to focus on the role of ion channels/transporters loss in the parietal cells induced gastric diffused mucosal injury.
Collapse
Affiliation(s)
- Yingying Zhao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zilin Deng
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Minglin Zhang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Hu Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Taolang Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| | - Xuemei Liu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| |
Collapse
|
2
|
Beltrán LR, Sterneder S, Hasural A, Paetz S, Hans J, Ley JP, Somoza V. Reducing the Bitter Taste of Pharmaceuticals Using Cell-Based Identification of Bitter-Masking Compounds. Pharmaceuticals (Basel) 2022; 15:ph15030317. [PMID: 35337115 PMCID: PMC8953435 DOI: 10.3390/ph15030317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/20/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
The palatability of a pharmaceutical preparation is a significant obstacle in developing a patient-friendly dosage form. Bitter taste is an important factor for patients in (i) selecting a certain drug from generic products available in the market and (ii) adhering to a therapeutic regimen. The various methods developed for identification of bitter tasting and bitter-taste modulating compounds present a number of limitations, ranging from limited sensitivity to lack of close correlations with sensory data. In this study, we demonstrate a fluorescence-based assay, analyzing the bitter receptor TAS2R-linked intracellular pH (pHi) of human gastric parietal (HGT-1) cells as a suitable tool for the identification of bitter tasting and bitter-taste modulating pharmaceutical compounds and preparations, which resembles bitter taste perception. Among the fluorometric protocols established to analyze pHi changes, one of the most commonly employed assays is based on the use of the pH-sensitive dye SNARF-1 AM. This methodology presents some limitations; over time, the assay shows a relatively low signal amplitude and sensitivity. Here, the SNARF-1 AM methodology was optimized. The identified bicarbonate extrusion mechanisms were partially inhibited, and measurements were carried out in a medium with lower intrinsic fluorescence, with no need for controlling external CO2 levels. We applied the assay for the screening of flavonoids as potential bitter-masking compounds for guaifenesin, a bitter-tasting antitussive drug. Our findings revealed that eriodictyol, hesperitin and phyllodulcin were the most potent suitable candidates for bitter-masking activity, verified in a human sensory trial.
Collapse
Affiliation(s)
- Leopoldo Raul Beltrán
- Department of Physiological Chemistry, University of Vienna, 1090 Vienna, Austria; (L.R.B.); (S.S.); (A.H.)
| | - Sonja Sterneder
- Department of Physiological Chemistry, University of Vienna, 1090 Vienna, Austria; (L.R.B.); (S.S.); (A.H.)
| | - Ayse Hasural
- Department of Physiological Chemistry, University of Vienna, 1090 Vienna, Austria; (L.R.B.); (S.S.); (A.H.)
| | - Susanne Paetz
- Symrise AG, Ingredient Research Flavor & Nutrition, 37603 Holzminden, Germany; (S.P.); (J.H.); (J.P.L.)
| | - Joachim Hans
- Symrise AG, Ingredient Research Flavor & Nutrition, 37603 Holzminden, Germany; (S.P.); (J.H.); (J.P.L.)
| | - Jakob Peter Ley
- Symrise AG, Ingredient Research Flavor & Nutrition, 37603 Holzminden, Germany; (S.P.); (J.H.); (J.P.L.)
| | - Veronika Somoza
- Department of Physiological Chemistry, University of Vienna, 1090 Vienna, Austria; (L.R.B.); (S.S.); (A.H.)
- Leibniz-Institute of Food Systems Biology at the Technical University of Munich, 85354 Freising, Germany
- Nutritional Systems Biology, Technical University of Munich, 85354 Freising, Germany
- Correspondence: ; Tel.: +43-1-4277-70601
| |
Collapse
|
3
|
Effect of Food and Dosing Regimen on Safety and Efficacy of Proton Pump Inhibitors Therapy-A Literature Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18073527. [PMID: 33805341 PMCID: PMC8036504 DOI: 10.3390/ijerph18073527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
Proton pump inhibitors (PPIs) are the first-choice drugs used to prevent and treat acid-related diseases. However, a lack of satisfactory response to the standard PPI dose ("PPI failure") is often reported, especially in patients with gastroesophageal reflux disease. Poor compliance seems to be one of the main causes of PPI failure; hence, it is crucial to gain knowledge on how to properly administer PPIs. In this review, we aimed to evaluate the effect of food, beverages, and dosing regimen on pharmacokinetics and pharmacodynamics of PPIs and to frame recommendations for healthcare professionals to improve both patient's counseling and compliance to treatment with PPIs. A total of 201 papers were identified following a literature search. After full-text evaluation, 64 studies were included in the review. Co-administration of PPIs with a meal may affect both their bioavailability and effectiveness; however, the influence of food depends on the type of drug and its formulation. Except for pantoprazole, PPIs can be administered in the morning or evening; however, morning intake generally provides better daytime control of gastric acidity. In most cases, the choice of the proper schedule of administration should be based on the patient's symptoms and individual dosing preferences.
Collapse
|
4
|
Li W, Gelsinger S, Edwards A, Riehle C, Koch D. Transcriptome analysis of rumen epithelium and meta-transcriptome analysis of rumen epimural microbial community in young calves with feed induced acidosis. Sci Rep 2019; 9:4744. [PMID: 30894588 PMCID: PMC6426933 DOI: 10.1038/s41598-019-40375-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/04/2019] [Indexed: 02/07/2023] Open
Abstract
Many common management practices used to raise dairy calves while on milk and during weaning can cause rumen acidosis. Ruminal pH has long been used to identify ruminal acidosis. However, few attempts were undertaken to understand the role of prolonged ruminal acidosis on rumen microbial community or host health in young calves long after weaning. Thus, the molecular changes associated with prolonged rumen acidosis in post weaning young calves are largely unknown. In this study, we induced ruminal acidosis by feeding a highly processed, starch-rich diet to calves starting from one week of age through 16 weeks. Rumen epithelial tissues were collected at necropsy at 17 weeks of age. Transcriptome analyses on the rumen epithelium and meta-transcriptome analysis of rumen epimural microbial communities were carried out. Calves with induced ruminal acidosis showed significantly less weight gain over the course of the experiment, in addition to substantially lower ruminal pH in comparison to the control group. For rumen epithelial transcriptome, a total of 672 genes (fold-change, FC ≥ 1.5; adjusted-p ≤ 0.05) showed significant differential expression in comparison to control. Biological pathways impacted by these differentially expressed genes included cell signaling and morphogenesis, indicating the impact of ruminal acidosis on rumen epithelium development. rRNA read-based microbial classification indicated significant increase in abundance of several genera in calves with induced acidosis. Our study provides insight into host rumen transcriptome changes associated with prolonged acidosis in post weaning calves. Shifts in microbial species abundance are promising for microbial species-based biomarker development and artificial manipulation. Such knowledge provides a foundation for future more precise diagnosis and preventative management of rumen acidosis in dairy calves.
Collapse
Affiliation(s)
- Wenli Li
- The Cell Wall Utilization and Biology Laboratory, US Dairy Forage Research Center, USDA ARS, Madison, WI, 53706, USA.
| | - Sonia Gelsinger
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Andrea Edwards
- The Cell Wall Utilization and Biology Laboratory, US Dairy Forage Research Center, USDA ARS, Madison, WI, 53706, USA
| | - Christina Riehle
- Department of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Daniel Koch
- Department of Computer Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
5
|
Gagic M, Jamroz E, Krizkova S, Milosavljevic V, Kopel P, Adam V. Current Trends in Detection of Histamine in Food and Beverages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:773-783. [PMID: 30585064 DOI: 10.1021/acs.jafc.8b05515] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Histamine is a heterocyclic amine formed by decarboxylation of the amino acid l-histidine. It is involved in the local regulation of physiological processes but also can occur exogenously in the food supply. Histamine is toxic at high intakes; therefore, determination of the histamine level in food is an important aspect of food safety. This article will review the current understanding of physiological functions of endogenous and ingested histamine with a particular focus placed on existing and emerging technologies for histamine quantification in food. Methods reported in this article are sequentially arranged and provide a brief overview of analytical methods reported, including those based on nanotechnologies.
Collapse
Affiliation(s)
- Milica Gagic
- Department of Chemistry and Biochemistry, Faculty of AgriSciences , Mendel University in Brno , Zemedelska 1 , CZ-613 00 Brno , Czech Republic
- Central European Institute of Technology , Brno University of Technology , Purkynova 123 , CZ-612 00 Brno , Czech Republic
| | - Ewelina Jamroz
- Institute of Chemistry , University of Agriculture in Cracow , Balicka Street 122 , PL-30-149 Cracow , Poland
| | - Sona Krizkova
- Department of Chemistry and Biochemistry, Faculty of AgriSciences , Mendel University in Brno , Zemedelska 1 , CZ-613 00 Brno , Czech Republic
- Central European Institute of Technology , Brno University of Technology , Purkynova 123 , CZ-612 00 Brno , Czech Republic
| | - Vedran Milosavljevic
- Department of Chemistry and Biochemistry, Faculty of AgriSciences , Mendel University in Brno , Zemedelska 1 , CZ-613 00 Brno , Czech Republic
- Central European Institute of Technology , Brno University of Technology , Purkynova 123 , CZ-612 00 Brno , Czech Republic
| | - Pavel Kopel
- Department of Chemistry and Biochemistry, Faculty of AgriSciences , Mendel University in Brno , Zemedelska 1 , CZ-613 00 Brno , Czech Republic
- Central European Institute of Technology , Brno University of Technology , Purkynova 123 , CZ-612 00 Brno , Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Faculty of AgriSciences , Mendel University in Brno , Zemedelska 1 , CZ-613 00 Brno , Czech Republic
- Central European Institute of Technology , Brno University of Technology , Purkynova 123 , CZ-612 00 Brno , Czech Republic
| |
Collapse
|
6
|
Sakai H, Fujii T, Takeguchi N. Proton-Potassium (H+/K+) ATPases: Properties and Roles in Health and Diseases. Met Ions Life Sci 2016; 16:459-83. [DOI: 10.1007/978-3-319-21756-7_13] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
7
|
Arin RM, Vallejo AI, Rueda Y, Fresnedo O, Ochoa B. Stimulation of gastric acid secretion by rabbit parietal cell A2B adenosine receptor activation. Am J Physiol Cell Physiol 2015; 309:C823-34. [DOI: 10.1152/ajpcell.00224.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/08/2015] [Indexed: 11/22/2022]
Abstract
Adenosine modulates different functional activities in many cells of the gastrointestinal tract; some of them are believed to be mediated by interaction with its four G protein-coupled receptors. The renewed interest in the adenosine A2B receptor (A2BR) subtype can be traced by studies in which the introduction of new genetic and chemical tools has widened the pharmacological and structural knowledge of this receptor as well as its potential therapeutic use in cancer and inflammation- or hypoxia-related pathologies. In the acid-secreting parietal cells of the gastric mucosa, the use of various radioligands for adenosine receptors suggested the presence of the A2 adenosine receptor subtype(s) on the cell surface. Recently, we confirmed A2BR expression in native, nontransformed parietal cells at rest by using flow cytometry and confocal microscopy. In this study, we show that A2BR is functional in primary rabbit gastric parietal cells, as indicated by the fact that agonist binding to A2BR increased adenylate cyclase activity and acid production. In addition, both acid production and radioligand binding of adenosine analogs to isolated cell membranes were potently blocked by selective A2BR antagonists, whereas ligands for A1, A2A, and A3 adenosine receptors failed to abolish activation. We conclude that rabbit gastric parietal cells possess functional A2BR proteins that are coupled to Gs and stimulate HCl production upon activation. Whether adenosine- and A2BR-mediated functional responses play a role in human gastric pathophysiology is yet to be elucidated.
Collapse
Affiliation(s)
- Rosa María Arin
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Ana Isabel Vallejo
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Yuri Rueda
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Olatz Fresnedo
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Begoña Ochoa
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, Leioa, Spain
| |
Collapse
|
8
|
Natarajan P, Crothers JM, Rosen JE, Nakada SL, Rakholia M, Okamoto CT, Forte JG, Machen TE. Myosin IIB and F-actin control apical vacuolar morphology and histamine-induced trafficking of H-K-ATPase-containing tubulovesicles in gastric parietal cells. Am J Physiol Gastrointest Liver Physiol 2014; 306:G699-710. [PMID: 24578340 PMCID: PMC3989701 DOI: 10.1152/ajpgi.00316.2013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Selective inhibitors of myosin or actin function and confocal microscopy were used to test the role of an actomyosin complex in controlling morphology, trafficking, and fusion of tubulovesicles (TV) containing H-K-ATPase with the apical secretory canaliculus (ASC) of primary-cultured rabbit gastric parietal cells. In resting cells, myosin IIB and IIC, ezrin, and F-actin were associated with ASC, whereas H-K-ATPase localized to intracellular TV. Histamine caused fusion of TV with ASC and subsequent expansion resulting from HCl and water secretion; F-actin and ezrin remained associated with ASC whereas myosin IIB and IIC appeared to dissociate from ASC and relocalize to the cytoplasm. ML-7 (inhibits myosin light chain kinase) caused ASC of resting cells to collapse and most myosin IIB, F-actin, and ezrin to dissociate from ASC. TV were unaffected by ML-7. Jasplakinolide (stabilizes F-actin) caused ASC to develop large blebs to which actin, myosin II, and ezrin, as well as tubulin, were prominently localized. When added prior to stimulation, ML-7 and jasplakinolide prevented normal histamine-stimulated transformations of ASC/TV and the cytoskeleton, but they did not affect cells that had been previously stimulated with histamine. These results indicate that dynamic pools of actomyosin are required for maintenance of ASC structure in resting cells and for trafficking of TV to ASC during histamine stimulation. However, the dynamic pools of actomyosin are not required once the histamine-stimulated transformation of TV/ASC and cytoskeleton has occurred. These results also show that vesicle trafficking in parietal cells shares mechanisms with similar processes in renal collecting duct cells, neuronal synapses, and skeletal muscle.
Collapse
Affiliation(s)
- Paramasivam Natarajan
- 1Department of Molecular and Cell Biology, University of California, Berkeley, California; and
| | - James M. Crothers
- 1Department of Molecular and Cell Biology, University of California, Berkeley, California; and
| | - Jared E. Rosen
- 1Department of Molecular and Cell Biology, University of California, Berkeley, California; and
| | - Stephanie L. Nakada
- 1Department of Molecular and Cell Biology, University of California, Berkeley, California; and
| | - Milap Rakholia
- 1Department of Molecular and Cell Biology, University of California, Berkeley, California; and
| | - Curtis T. Okamoto
- 2Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - John G. Forte
- 1Department of Molecular and Cell Biology, University of California, Berkeley, California; and
| | - Terry E. Machen
- 1Department of Molecular and Cell Biology, University of California, Berkeley, California; and
| |
Collapse
|
9
|
Barrera M, Bahamondes V, Sepúlveda D, Quest A, Castro I, Cortés J, Aguilera S, Urzúa U, Molina C, Pérez P, Ewert P, Alliende C, Hermoso M, González S, Leyton C, González M. Sjögren's syndrome and the epithelial target: A comprehensive review. J Autoimmun 2013; 42:7-18. [DOI: 10.1016/j.jaut.2013.02.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 02/11/2013] [Indexed: 12/12/2022]
|
10
|
Nguyen N, Kozer-Gorevich N, Gliddon BL, Smolka AJ, Clayton AH, Gleeson PA, van Driel IR. Independent trafficking of the KCNQ1 K+ channel and H+-K+-ATPase in gastric parietal cells from mice. Am J Physiol Gastrointest Liver Physiol 2013; 304:G157-66. [PMID: 23154976 DOI: 10.1152/ajpgi.00346.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastric acid secretion by the H(+)-K(+)-ATPase at the apical surface of activated parietal cells requires luminal K(+) provided by the KCNQ1/KCNE2 K(+) channel. However, little is known about the trafficking and relative spatial distribution of KCNQ1 and H(+)-K(+)-ATPase in resting and activated parietal cells and the capacity of KCNQ1 to control acid secretion. Here we show that inhibition of KCNQ1 activity quickly curtails gastric acid secretion in vivo, even when the H(+)-K(+)-ATPase is permanently anchored in the apical membrane, demonstrating a key role of the K(+) channel in controlling acid secretion. Three-dimensional imaging analysis of isolated mouse gastric units revealed that the majority of KCNQ1 resides in an intracytoplasmic, Rab11-positive compartment in resting parietal cells, distinct from H(+)-K(+)-ATPase-enriched tubulovesicles. Upon activation, there was a significant redistribution of H(+)-K(+)-ATPase and KCNQ1 from intracytoplasmic compartments to the apical secretory canaliculi. Significantly, high Förster resonance energy transfer was detected between H(+)-K(+)-ATPase and KCNQ1 in activated, but not resting, parietal cells. These findings demonstrate that H(+)-K(+)-ATPase and KCNQ1 reside in independent intracytoplasmic membrane compartments, or membrane domains, and upon activation of parietal cells, both membrane proteins are transported, possibly via Rab11-positive recycling endosomes, to apical membranes, where the two molecules are closely physically opposed. In addition, these studies indicate that acid secretion is regulated by independent trafficking of KCNQ1 and H(+)-K(+)-ATPase.
Collapse
Affiliation(s)
- Nhung Nguyen
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Australia
| | | | | | | | | | | | | |
Collapse
|
11
|
Fujii T, Fujita K, Takeguchi N, Sakai H. Function of K⁺-Cl⁻ cotransporters in the acid secretory mechanism of gastric parietal cells. Biol Pharm Bull 2011; 34:810-2. [PMID: 21628876 DOI: 10.1248/bpb.34.810] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Gastric proton pump (H⁺, K⁺-ATPase) secretes H⁺ of acid (HCl) via the luminal membrane of parietal cells. For the HCl secretion, Cl⁻- and K⁺-transporting proteins are required. Recent our studies have demonstrated that K⁺-Cl⁻ cotransporters (KCC3a and KCC4) are expressed in gastric parietal cells. KCC3a is associated with Na⁺, K⁺-ATPase in the basolateral membrane, and KCC4 is associated with H⁺, K⁺-ATPase in the apical canalicular membrane. This paper summarizes the functional association between KCCs and P-type ATPases and the contribution of these complexes to acid secretion in gastric parietal cells.
Collapse
Affiliation(s)
- Takuto Fujii
- Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
| | | | | | | |
Collapse
|
12
|
Matsukawa J, Hori Y, Nishida H, Kajino M, Inatomi N. A comparative study on the modes of action of TAK-438, a novel potassium-competitive acid blocker, and lansoprazole in primary cultured rabbit gastric glands. Biochem Pharmacol 2011; 81:1145-51. [DOI: 10.1016/j.bcp.2011.02.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 02/14/2011] [Accepted: 02/15/2011] [Indexed: 02/06/2023]
|
13
|
Grainger S, Savory JGA, Lohnes D. Cdx2 regulates patterning of the intestinal epithelium. Dev Biol 2010; 339:155-65. [PMID: 20043902 DOI: 10.1016/j.ydbio.2009.12.025] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 12/16/2009] [Accepted: 12/17/2009] [Indexed: 12/25/2022]
Abstract
Cdx1, Cdx2 and Cdx4 encode homeodomain transcription factors that are involved in vertebral anterior-posterior (AP) patterning. Cdx1 and Cdx2 are also expressed in the intestinal epithelium during development, suggesting a role in this tissue. Intestinal defects have not been reported in Cdx1 null mutants, while Cdx2 null mutants die at embryonic day 3.5 (E3.5), thus precluding assessment of the null phenotype at later stages. To circumvent this latter shortcoming, we have used a conditional Cre-lox strategy to inactivate Cdx2 in the intestinal epithelium. Using this approach, we found that ablation of Cdx2 at E13.5 led to a transformation of the small intestine to a pyloric stomach-like identity, although the molecular nature of the underlying mesenchyme remained unchanged. Further analysis of Cdx1-Cdx2 double mutants suggests that Cdx1 does not play a critical role in the development of the small intestine, at least after E13.5.
Collapse
Affiliation(s)
- Stephanie Grainger
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
14
|
Abstract
The parietal cell is responsible for secreting concentrated hydrochloric acid into the gastric lumen. To fulfill this task, it is equipped with a broad variety of functionally coupled apical and basolateral ion transport proteins. The concerted scientific effort over the last years by a variety of researchers has provided us with the molecular identity of many of these transport mechanisms, thereby contributing to the clarification of persistent controversies in the field. This article will briefly review the current model of parietal cell physiology and ion transport in particular and will update the existing models of apical and basolateral transport in the parietal cell.
Collapse
Affiliation(s)
- Sascha Kopic
- Department of Surgery, Yale University, School of Medicine, New Haven, Connecticut
| | - Michael Murek
- Department of Surgery, Yale University, School of Medicine, New Haven, Connecticut
| | - John P. Geibel
- Department of Surgery, Yale University, School of Medicine, New Haven, Connecticut
| |
Collapse
|
15
|
Affiliation(s)
- Eric S. Fortune
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Maurice J. Chacron
- Department of Physiology, Center for Nonlinear Dynamics, McGill University, Montreal, Canada
- Department of Physics, McGill University, Montreal, Canada
- * E-mail:
| |
Collapse
|
16
|
Zhang H, Chen X, Bollag WB, Bollag RJ, Sheehan DJ, Chew CS. Lasp1 gene disruption is linked to enhanced cell migration and tumor formation. Physiol Genomics 2009; 38:372-85. [PMID: 19531578 DOI: 10.1152/physiolgenomics.00048.2009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Lasp1 is an actin-binding, signaling pathway-regulated phosphoprotein that is overexpressed in several cancers. siRNA knockdown in cell lines retards cell migration, suggesting the possibility that Lasp1 upregulation influences cancer metastasis. Herein, we utilized a recently developed gene knockout model to assess the role of Lasp1 in modulating nontransformed cell functions. Wound healing and tumor initiation progressed more rapidly in Lasp1(-/-) mice compared with Lasp1(+/+) controls. Embryonic fibroblasts (MEFs) derived from Lasp1(-/-) mice also migrated more rapidly in vitro. These MEFs characteristically possessed increased focal adhesion numbers and displayed more rapid attachment compared with wild-type MEFs. Differential microarray analyses revealed alterations in message expression for proteins implicated in cell migration, adhesion, and cytoskeletal organization. Notably, the focal adhesion protein, lipoma preferred partner (LPP), a zyxin family member and putative Lasp1 binding protein, was increased about twofold. Because LPP gene disruption reduces cell migration, we hypothesize that LPP plays a role in enhancing the migratory capacity of Lasp1(-/-) MEFs, perhaps by modifying the subcellular localization of other motility-associated proteins. The striking contrast in the functional effects of loss of Lasp1 in innate cells compared with cell lines reveals distinct differences in mechanisms of motility and attachment in these models.
Collapse
Affiliation(s)
- Han Zhang
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia, USA
| | | | | | | | | | | |
Collapse
|
17
|
Fujii T, Takahashi Y, Ikari A, Morii M, Tabuchi Y, Tsukada K, Takeguchi N, Sakai H. Functional Association between K+-Cl- Cotransporter-4 and H+,K+-ATPase in the Apical Canalicular Membrane of Gastric Parietal Cells. J Biol Chem 2009; 284:619-629. [DOI: 10.1074/jbc.m806562200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
18
|
Perez-Zoghbi JF, Mayora A, Ruiz MC, Michelangeli F. Heterogeneity of acid secretion induced by carbachol and histamine along the gastric gland axis and its relationship to [Ca2+]i. Am J Physiol Gastrointest Liver Physiol 2008; 295:G671-81. [PMID: 18669625 DOI: 10.1152/ajpgi.90224.2008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gastric glands of the mammalian fundic mucosa are constituted by different cell types. Gastric fluid is a mixture of acid, alkali, ions, enzymes, and mucins secreted by parietal, chief, and mucous cells. We studied activation of acid secretion using LysoSensor Yellow/Blue in conjunction with fluo 3 to measure changes in pH and Ca(2+) in isolated rabbit gastric glands. We evidenced a spatial heterogeneity in the amplitude of acid response along the gland axis under histamine and cholinergic stimulation. Carbachol induced a transitory pH increase before acidification. This relative alkalinization may be related to granule release from other cell types. Omeprazole inhibited the acid component but not the rise in pH. Histamine stimulated acid secretion without increase of lumen pH. We studied the relationship between Ca(2+) release and/or entry and H(+) secretion in glands stimulated by carbachol. Ca(2+) release was associated with a fast and transient components of H(+) secretion. We found a linear relationship between Ca(2+) release and H(+) secretion. Ca(2+) entry was associated with a second slow and larger component of acid secretion. The fast component may be the result of activation of Cl(-) and K(+) channels and hence H(+)/K(+) pumps already present in the membrane, whereas the slow component might be associated with translocation of H(+)/K(+) pumps to the canaliculi. In conclusion, with cholinergic stimulation, gastric glands secrete a mixture of acid and other product(s) with a pH above 4.2, both triggered by Ca(2+) release. Maintenance of acid secretion depends on Ca(2+) entry and perhaps membrane fusion.
Collapse
|
19
|
Jain RN, Al-Menhali AA, Keeley TM, Ren J, El-Zaatari M, Chen X, Merchant JL, Ross TS, Chew CS, Samuelson LC. Hip1r is expressed in gastric parietal cells and is required for tubulovesicle formation and cell survival in mice. J Clin Invest 2008; 118:2459-70. [PMID: 18535670 DOI: 10.1172/jci33569] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 04/30/2008] [Indexed: 11/17/2022] Open
Abstract
Huntingtin interacting protein 1 related (Hip1r) is an F-actin- and clathrin-binding protein involved in vesicular trafficking. In this study, we demonstrate that Hip1r is abundantly expressed in the gastric parietal cell, predominantly localizing with F-actin to canalicular membranes. Hip1r may provide a critical function in vivo, as demonstrated by extensive changes to parietal cells and the gastric epithelium in Hip1r-deficient mice. Electron microscopy revealed abnormal apical canalicular membranes and loss of tubulovesicles in mutant parietal cells, suggesting that Hip1r is necessary for the normal trafficking of these secretory membranes. Accordingly, acid secretory dynamics were altered in mutant parietal cells, with enhanced activation and acid trapping, as measured in isolated gastric glands. At the whole-organ level, gastric acidity was reduced in Hip1r-deficient mice, and the gastric mucosa was grossly transformed, with fewer parietal cells due to enhanced apoptotic cell death and glandular hypertrophy associated with cellular transformation. Hip1r-deficient mice had increased expression of the gastric growth factor gastrin, and mice mutant for both gastrin and Hip1r exhibited normalization of both proliferation and gland height. Taken together, these studies demonstrate that Hip1r plays a significant role in gastric physiology, mucosal architecture, and secretory membrane dynamics in parietal cells.
Collapse
Affiliation(s)
- Renu N Jain
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Heitzmann D, Warth R. Physiology and pathophysiology of potassium channels in gastrointestinal epithelia. Physiol Rev 2008; 88:1119-82. [PMID: 18626068 DOI: 10.1152/physrev.00020.2007] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Epithelial cells of the gastrointestinal tract are an important barrier between the "milieu interne" and the luminal content of the gut. They perform transport of nutrients, salts, and water, which is essential for the maintenance of body homeostasis. In these epithelia, a variety of K(+) channels are expressed, allowing adaptation to different needs. This review provides an overview of the current literature that has led to a better understanding of the multifaceted function of gastrointestinal K(+) channels, thereby shedding light on pathophysiological implications of impaired channel function. For instance, in gastric mucosa, K(+) channel function is a prerequisite for acid secretion of parietal cells. In epithelial cells of small intestine, K(+) channels provide the driving force for electrogenic transport processes across the plasma membrane, and they are involved in cell volume regulation. Fine tuning of salt and water transport and of K(+) homeostasis occurs in colonic epithelia cells, where K(+) channels are involved in secretory and reabsorptive processes. Furthermore, there is growing evidence for changes in epithelial K(+) channel expression during cell proliferation, differentiation, apoptosis, and, under pathological conditions, carcinogenesis. In the future, integrative approaches using functional and postgenomic/proteomic techniques will help us to gain comprehensive insights into the role of K(+) channels of the gastrointestinal tract.
Collapse
Affiliation(s)
- Dirk Heitzmann
- Institute of Physiology and Clinic and Policlinic for Internal Medicine II, Regensburg, Germany
| | | |
Collapse
|
21
|
Gliddon BL, Nguyen NV, Gunn PA, Gleeson PA, van Driel IR. Isolation, culture and adenoviral transduction of parietal cells from mouse gastric mucosa. Biomed Mater 2008; 3:034117. [DOI: 10.1088/1748-6041/3/3/034117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
22
|
Chew CS, Chen X, Bollag RJ, Isales C, Ding KH, Zhang H. Targeted disruption of the Lasp-1 gene is linked to increases in histamine-stimulated gastric HCl secretion. Am J Physiol Gastrointest Liver Physiol 2008; 295:G37-G44. [PMID: 18483181 PMCID: PMC2494726 DOI: 10.1152/ajpgi.90247.2008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Accepted: 05/08/2008] [Indexed: 01/31/2023]
Abstract
Lasp-1 (LIM and SH3 domain protein 1) is a multidomain actin-binding protein that is differentially expressed within epithelial tissues and brain. In the gastric mucosa, Lasp-1 is highly expressed in the HCl-secreting parietal cell, where it is prominently localized within the F-actin-rich subcellular regions. Histamine-induced elevation of parietal cell [cAMP]i increases Lasp-1 phosphorylation, which is correlated with activation of HCl secretion. To determine whether Lasp-1 is involved in the regulation of HCl secretion in vivo, we generated a murine model with a targeted disruption of the Lasp-1 gene. Lasp-1-null mice had slightly lower body weights but developed normally and had no overt phenotypic abnormalities. Basal HCl secretion was unaffected by loss of Lasp-1, but histamine stimulation induced a more robust acid secretory response in Lasp-1-null mice compared with wild-type littermates. A similar effect of histamine was observed in isolated gastric glands on the basis of measurements of accumulation of the weak base [14C]aminopyrine. In addition, inhibition of the acid secretory response to histamine by H2 receptor blockade with ranitidine proceeded more slowly in glands from Lasp-1-null mice. These findings support the conclusion that Lasp-1 is involved in the regulation of parietal HCl secretion. We speculate that cAMP-dependent phosphorylation of Lasp-1 alters interactions with F-actin and/or endocytic proteins that interact with Lasp-1, thereby regulating the trafficking/activation of the H+, K+-ATPase (proton pump).
Collapse
Affiliation(s)
- Catherine S Chew
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, GA 30912-3175, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Pagliocca A, Hegyi P, Venglovecz V, Rackstraw SA, Khan Z, Burdyga G, Wang TC, Dimaline R, Varro A, Dockray GJ. Identification of ezrin as a target of gastrin in immature mouse gastric parietal cells. Exp Physiol 2008; 93:1174-89. [PMID: 18567601 DOI: 10.1113/expphysiol.2008.042648] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The gastric acid-secreting parietal cell exhibits profound morphological changes on stimulation. Studies in gastrin null (Gas-KO) mice indicate that maturation of parietal cell function depends on the hormone gastrin acting at the G-protein-coupled cholecystokinin 2 receptor. The relevant cellular mechanisms are unknown. The application of differential mRNA display to samples of the gastric corpus of wild-type (C57BL/6) and Gas-KO mice identified the cytoskeletal linker protein, ezrin, as a previously unsuspected target of gastrin. Gastrin administered in vivo or added to gastric glands in vitro increased ezrin abundance in Gas-KO parietal cells. In parietal cells of cultured gastric glands from wild-type mice treated with gastrin, histamine or carbachol, ezrin was localized to vesicular structures resembling secretory canaliculi. In contrast, in cultured parietal cells from Gas-KO mice, ezrin was typically distributed in the cytosol, and this did not change after incubation with gastrin, histamine or carbachol. However, priming with gastrin for approximately 24 h, either in vivo prior to cell culture or by addition to cultured gastric glands, induced the capacity for secretagogue-stimulated localization of ezrin to large vesicular structures in Gas-KO mice. Similarly, in a functional assay based on measurement of intracellular pH, cultured parietal cells from Gas-KO mice were refractory to gastrin unless primed. The priming effect of gastrin was not attributable to the paracrine mediator histamine, but was prevented by inhibitors of protein kinase C and transactivation of the epidermal growth factor receptor. We conclude that in gastrin null mice there is reduced ezrin expression and a defect in ezrin subcellular distribution in gastric parietal cells, and that both can be reversed by priming with gastrin.
Collapse
Affiliation(s)
- Adelina Pagliocca
- Physiological Laboratory, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Heitzmann D, Warth R. No potassium, no acid: K+ channels and gastric acid secretion. Physiology (Bethesda) 2008; 22:335-41. [PMID: 17928547 DOI: 10.1152/physiol.00016.2007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The gastric H+-K+-ATPase pumps H+ into the lumen and takes up K+ in parallel. In the acid-producing parietal cells, luminal KCNE2/KCNQ1 K+ channels play a pivotal role in replenishing K+ in the luminal fluid. Inactivation of KCNE2/KCNQ1 channels abrogates gastric acid secretion and dramatically modifies the architecture of gastric mucosa.
Collapse
|
25
|
Schreiber S, Garten D, Nguyen TH, Konradt M, Bücker R, Scheid P. In situ measurement of pH in the secreting canaliculus of the gastric parietal cell and adjacent structures. Cell Tissue Res 2007; 329:313-20. [PMID: 17505843 DOI: 10.1007/s00441-007-0427-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Accepted: 04/13/2007] [Indexed: 01/29/2023]
Abstract
The gastric H(+)/K(+)-ATPase is located within an infolding (secretory canaliculus) of the apical plasma membrane of gastric parietal cells. Our aim was to measure the pH values in the cytosol and canaliculus of the acid-secreting parietal cell and the adjacent gland lumen in situ. We used ultrafine double-barreled tip-sealed microelectrodes at high acceleration rates for intracellular and canalicular measurements. Immunohistochemical staining of the parietal cells was used to identify the track of the electrode and to estimate the position of the electrode tip at the time of the last intracellular measurement. En route to the deepest regions of the mucosa, where the average gland lumen pH was approximately 3, and on advancing in steps of 2 mum, the electrode entered the cytosol of the parietal cells, where the pH value was 7.4. Advancing the electrode further resulted, in several instances, in a sharp decrease in pH to an average value of 1.7 +/- 0.2, which we interpreted as the measurement within the canaliculus. When the electrode was advanced even further, the pH reading returned to the cytosolic value. From the difference in pH between the secreting canaliculus and the adjacent gland lumen, we concluded that the released acid was immediately buffered. Thus, the only cellular structure directly exposed to the highly acidic canalicular content is the apical membrane forming the canaliculus in the parietal cell.
Collapse
Affiliation(s)
- Sören Schreiber
- Institut für Physiologie (MA 2/149), Ruhr-Universität Bochum, Universitätsstrasse 150, 44801, Bochum, Germany.
| | | | | | | | | | | |
Collapse
|
26
|
Fu D, Roufogalis BD. Actin disruption inhibits endosomal traffic of P-glycoprotein-EGFP and resistance to daunorubicin accumulation. Am J Physiol Cell Physiol 2007; 292:C1543-52. [PMID: 17122416 DOI: 10.1152/ajpcell.00068.2006] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Intracellular traffic of human P-glycoprotein (P-gp), a membrane transporter responsible for multidrug resistance in cancer chemotherapy, was investigated using a P-gp and enhanced green fluorescent fusion protein (P-gp-EGFP) in human breast cancer MCF-7 cells. The stably expressed P-gp-EGFP from a clonal cell population was functional as a drug efflux pump, as demonstrated by the inhibition of daunorubicin accumulation and the conferring of resistance of the cells to colchicine and daunorubicin. Colocalization experiments demonstrated that a small fraction of the total P-gp-EGFP expressed was localized intracellularly and was present in early endosome and lysosome compartments. P-gp-EGFP traffic was shown to occur via early endosome transport to the plasma membrane. Subsequent movement of P-gp-EGFP away from the plasma membrane occurred by endocytosis to the early endosome and lysosome. The component of the cytoskeleton responsible for P-gp-EGFP traffic was demonstrated to be actin rather than microtubules. In functional studies it was shown that in parallel with the interruption of the traffic of P-gp-EGFP, cellular accumulation of the P-gp substrate daunorubicin was increased after cells were treated with actin inhibitors, and cell proliferation was inhibited to a greater extent than in the presence of daunorubicin alone. The actin dependence of P-gp traffic and the parallel changes in cytotoxic drug accumulation demonstrated in this study delineates the pathways of P-gp traffic and may provide a new approach to overcoming multidrug resistance in cancer chemotherapy.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Actins/metabolism
- Antibiotics, Antineoplastic/metabolism
- Antibiotics, Antineoplastic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Line, Tumor
- Cell Membrane/metabolism
- Cell Proliferation/drug effects
- Colchicine/pharmacology
- Cycloheximide/pharmacology
- Cytochalasin D/pharmacology
- Daunorubicin/metabolism
- Daunorubicin/pharmacology
- Drug Resistance, Multiple
- Drug Resistance, Neoplasm
- Endosomes/metabolism
- Endosomes/ultrastructure
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Humans
- Lysosomes/metabolism
- Lysosomes/ultrastructure
- Microtubules/metabolism
- Protein Synthesis Inhibitors/pharmacology
- Protein Transport
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Thiazolidines/pharmacology
Collapse
Affiliation(s)
- Dong Fu
- Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | | |
Collapse
|
27
|
Remy C, Kirchhoff P, Hafner P, Busque SM, Müeller MK, Geibel JP, Wagner CA. Stimulatory Pathways of the Calcium-Sensing Receptor on Acid Secretion in Freshly Isolated Human Gastric Glands. Cell Physiol Biochem 2007; 19:33-42. [PMID: 17310098 DOI: 10.1159/000099190] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2006] [Indexed: 11/19/2022] Open
Abstract
Gastric acid secretion is not only stimulated via the classical known neuronal and hormonal pathways but also by the Ca(2+)-Sensing Receptor (CaSR) located at the basolateral membrane of the acid-secretory gastric parietal cell. Stimulation of CaSR with divalent cations or the potent agonist Gd(3+) leads to activation of the H(+)/K(+)-ATPase and subsequently to gastric acid secretion. Here we investigated the intracellular mechanism(s) mediating the effects of the CaSR on H(+)/K(+)-ATPase activity in freshly isolated human gastric glands. Inhibition of heterotrimeric G-proteins (G(i) and G(o)) with pertussis toxin during stimulation of the CaSR with Gd(3+) only partly reduced the observed stimulatory effect. A similar effect was observed with the PLC inhibitor U73122. The reduction of the H(+)/K(+)-ATPase activity measured after incubation of gastric glands with BAPTA-AM, a chelator of intracellular Ca(2+), showed that intracellular Ca(2+) plays an important role in the signalling cascade. TMB-8, a ER Ca(2+)store release inhibitor, prevented the stimulation of H(+)/K(+)-ATPase activity. Also verapamil, an inhibitor of L-type Ca(2+)-channels reduced stimulation suggesting that both the release of intracellular Ca(2+) from the ER as well as Ca(2+) influx into the cell are involved in CaSR-mediated H(+)/K(+)-ATPase activation. Chelerythrine, a general inhibitor of protein kinase C, and Go 6976 which selectively inhibits Ca(2+)-dependent PKC(alpha) and PKC(betaI)-isozymes completely abolished the stimulatory effect of Gd(3+). In contrast, Ro 31-8220, a selective inhibitor of the Ca(2+)-independent PKCepsilon and PKC-delta isoforms reduced the stimulatory effect of Gd(3+) only about 60 %. On the other hand, activation of PKC with DOG led to an activation of H(+)/K(+)-ATPase activity which was only about 60 % of the effect observed with Gd(3+). Incubation of the parietal cells with PD 098059 to inhibit ERK1/2 MAP-kinases showed a significant reduction of the Gd(3+) effect. Thus, in the human gastric parietal cell the CaSR is coupled to pertussis toxin sensitive heterotrimeric G-Proteins and requires calcium to enhance the activity of the proton-pump. PLC, ERK 1/2 MAP-kinases as well as Ca(2+) dependent and Ca(2+)-independent PKC isoforms are part of the down-stream signalling cascade.
Collapse
Affiliation(s)
- Christine Remy
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zürich, Schweiz
| | | | | | | | | | | | | |
Collapse
|
28
|
Dufner MM, Kirchhoff P, Remy C, Hafner P, Müller MK, Cheng SX, Tang LQ, Hebert SC, Geibel JP, Wagner CA. The calcium-sensing receptor acts as a modulator of gastric acid secretion in freshly isolated human gastric glands. Am J Physiol Gastrointest Liver Physiol 2005; 289:G1084-90. [PMID: 16109841 DOI: 10.1152/ajpgi.00571.2004] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastric acid secretion is activated by two distinct pathways: a neuronal pathway via the vagus nerve and release of acetylcholine and an endocrine pathway involving gastrin and histamine. Recently, we demonstrated that activation of H(+)-K(+)-ATPase activity in parietal cells in freshly isolated rat gastric glands is modulated by the calcium-sensing receptor (CaSR). Here, we investigated if the CaSR is functionally expressed in freshly isolated gastric glands from human patients undergoing surgery and if the CaSR is influencing histamine-induced activation of H(+)-K(+)-ATPase activity. In tissue samples obtained from patients, immunohistochemistry demonstrated the expression in parietal cells of both subunits of gastric H(+)-K(+)-ATPase and the CaSR. Functional experiments using the pH-sensitive dye 2',7'-bis-(2-carboxyethyl)-5-(and 6)-carboxyfluorescein and measurement of intracellular pH changes allowed us to estimate the activity of H(+)-K(+)-ATPase in single freshly isolated human gastric glands. Under control conditions, H(+)-K(+)-ATPase activity was stimulated by histamine (100 microM) and inhibited by omeprazole (100 microM). Reduction of the extracellular divalent cation concentration (0 Mg(2+), 100 microM Ca(2+)) inactivated the CaSR and reduced histamine-induced activation of H(+)-K(+)-ATPase activity. In contrast, activation of the CaSR with the trivalent cation Gd(3+) caused activation of omeprazole-sensitive H(+)-K(+)-ATPase activity even in the absence of histamine and under conditions of low extracellular divalent cations. This stimulation was not due to release of histamine from neighbouring enterochromaffin-like cells as the stimulation persisted in the presence of the H(2) receptor antagonist cimetidine (100 microM). Furthermore, intracellular calcium measurements with fura-2 and fluo-4 showed that activation of the CaSR by Gd(3+) led to a sustained increase in intracellular Ca(2+) even under conditions of low extracellular divalent cations. These experiments demonstrate the presence of a functional CaSR in the human stomach and show that this receptor may modulate the activity of acid-secreting H(+)-K(+)-ATPase in parietal cells. Furthermore, our results show the viability of freshly isolated human gastric glands and may allow the use of this preparation for experiments investigating the physiological regulation and properties of human gastric glands in vitro.
Collapse
|
29
|
Chew CS, Okamoto CT, Chen X, Thomas R. Drebrin E2 is differentially expressed and phosphorylated in parietal cells in the gastric mucosa. Am J Physiol Gastrointest Liver Physiol 2005; 289:G320-31. [PMID: 15790763 DOI: 10.1152/ajpgi.00002.2005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Developmentally regulated brain proteins (drebrins) are highly expressed in brain where they may regulate actin filament formation in dendritic spines. Recently, the drebrin E2 isoform was detected in certain epithelial cell types including the gastric parietal cell. In gastric parietal cells, activation of HCl secretion is correlated with actin filament formation and elongation within intracellular canaliculi, which are the sites of acid secretion. The aim of this study was to define the pattern of drebrin expression in gland units in the intact rabbit oxyntic gastric mucosa and to initiate approaches to define the functions of this protein in parietal cells. Drebrin E2 expression was limited entirely or almost entirely to parietal cells and depended upon the localization of parietal cells along the gland axis. Rabbit drebrin E2 was cloned and found to share 86% identity with human drebrin 1a and to possess a number of cross-species conserved protein-protein interaction and phosphorylation consensus sites. Two-dimensional Western blot and phosphoaffinity column analyses confirmed that drebrin is phosphorylated in parietal cells, and several candidate phosphorylation sites were identified by mass spectrometry. Overexpression of epitope-tagged drebrin E2 led to the formation of microspikes and F-actin-rich ring-like structures in cultured parietal cells and suppressed cAMP-dependent acid secretory responses. In Madin-Darby canine kidney cells, coexpression of epitope-tagged drebrin and the Rho family GTPase Cdc42, which induces filopodial extension, produced an additive increase in the length of microspike projections. Coexpression of dominant negative Cdc42 with drebrin E2 did not prevent drebrin-induced microspike formation. These findings suggest that 1) drebrin can induce the formation of F-actin-rich membrane projections by Cdc42-dependent and -independent mechanisms; and that 2) drebrin plays an active role in directing the secretagogue-dependent formation of F-actin-rich filaments on the parietal cell canalicular membrane. Finally, the differential distribution of drebrin in parietal cells along the gland axis suggests that drebrin E2 may be an important marker of parietal cell differentiation and functionality.
Collapse
Affiliation(s)
- Catherine S Chew
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Sanders R&E Bldg., Rm. CB 2803, Augusta, GA, USA.
| | | | | | | |
Collapse
|
30
|
Affiliation(s)
- Raúl A Marinelli
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina.
| | | | | |
Collapse
|
31
|
Abstract
Uncoupling protein(UCP)1 is an integral membrane protein that is located in the mitochondrial inner membrane of brown adipocytes. Its physiological role is to mediate a regulated, thermogenic proton leak. UCP2 and UCP3 are recently identified UCP1 homologues. They also mediate regulated proton leak, and might function to control the production of superoxide and other downstream reactive oxygen species. However, their role in normal physiology remains unknown. Recent studies have shown that UCP2 has an important part in the pathogenesis of type-2 diabetes. The obscure roles of the UCP homologues in normal physiology, together with their emerging role in pathophysiology, provide exciting potential for further investigation.
Collapse
Affiliation(s)
- Evelina Chieregatti
- Department of Neuroscience, Vita-Salute San Raffaele University, Center of Excellence in Cellular Pathophysiology, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | | |
Collapse
|
32
|
Chew CS, Okamoto CT, Chen X, Qin HY. IQGAPs are differentially expressed and regulated in polarized gastric epithelial cells. Am J Physiol Gastrointest Liver Physiol 2005; 288:G376-87. [PMID: 15458922 DOI: 10.1152/ajpgi.00290.2004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
IQGAPs, GTPase-activating proteins with an IQ motif, are thought to regulate many actin cytoskeleton-based activities through interactions with Cdc42 and Rac. Recently, Cdc42 was implicated in regulation of gastric parietal cell HCl secretion, and IQGAP2 was immunolocalized with Cdc42 to F-actin-rich intracellular canalicular membranes of isolated gastric parietal cells in primary culture. Here we sought to define distribution and localization of IQGAP1 and IQGAP2 in major oxyntic (acid-secreting) gastric mucosal cell types and to determine whether secretory agonists modulate these proteins. Differential staining protocols were used to identify different cell populations (parietal, chief, surface/pit, and mucous neck cells) in semi-intact glands isolated from rabbit gastric mucosae and to characterize these same cells after dispersion and fractionation on isopycnic density gradients with simultaneous staining for F-actin, H+-K+-ATPase, and GSII lectin-binding sites. There was a pronounced increase in intracellular F-actin staining in dispersed chief cells, apparently from internalization of F-actin-rich apical membranes that normally abut the gland lumen. Therefore, other membrane-associated proteins might also be redistributed by disruption of cell-cell contacts. Western blot analyses were used to quantitate relative concentrations of IQGAPs in defined mucosal cell fractions, and gastric glands were used for in situ localizations. We detected uniform levels of IQGAP2 expression in oxyntic mucosal cells with predominant targeting to regions of cell-cell contact and nuclei of all cell types. IQGAP2 was not detected in parietal cell intracellular canaliculi. IQGAP1 expression was variable and targeted predominantly to the cortex of chief and mucous neck cells. Parietal cells expressed little or no IQGAP1 vs. other mucosal cell types. Phosphoprotein affinity chromatography, isoelectric focusing, and phosphorylation site analyses indicated that both IQGAP1 and IQGAP2 are phosphoproteins potentially regulated by [Ca2+]i/PKC and cAMP signaling pathways, respectively. Stimulation of glands with carbachol, which elevates [Ca2+]i and activates PKC, induced apparent translocation of IQGAP1, but not IQGAP2, to apical poles of chief (zymogen) and mucous neck cells. This response was mimicked by PMA but not by ionomycin or by elevation of [cAMP]i with forskolin. Our observations support a novel, PKC-dependent role for IQGAP1 in regulated exocytosis and suggest that IQGAP2 may play a more general role in regulating cell-cell interactions and possibly migration within the gastric mucosa.
Collapse
Affiliation(s)
- Catherine S Chew
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, GA 30912-3175, USA.
| | | | | | | |
Collapse
|
33
|
Abstract
As thousands of new genes are identified in genomics efforts, the rush is on to learn something about the functional roles of the proteins encoded by those genes. Clues to protein functions, activation states and protein-protein interactions have been revealed in focused studies of protein localization. With technical breakthroughs such as GFP protein tagging and recombinase cloning systems, large-scale screens of protein localization are now being undertaken.
Collapse
Affiliation(s)
- Nancy A O'Rourke
- Alliance for Cell Signaling, Microscopy Lab, Stanford University School of Medicine, 975 California Ave, Palo Alto, CA 94304, USA
| | | | | |
Collapse
|
34
|
Kato Y, Fukamachi H, Takano-Maruyama M, Aoe T, Murahashi Y, Horie S, Suzuki Y, Saito Y, Koseki H, Ohno H. Reduction of SNAP25 in acid secretion defect of Foxl1-/- gastric parietal cells. Biochem Biophys Res Commun 2004; 320:766-72. [PMID: 15240114 DOI: 10.1016/j.bbrc.2004.05.209] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2004] [Indexed: 10/26/2022]
Abstract
Foxl1 is a winged helix transcription factor expressed in the mesenchyme of the gastrointestinal tract. In the absence of Foxl1, parietal cells fail to secrete gastric acid in response to various secretagogue stimuli including cAMP. A marked decrease in H+,K(+)-ATPase expression was observed even though a substantial number of parietal cells still existed in Foxl1-deficient mice. Ultrastructural analysis suggested that the gastric acid secretion defect in Foxl1-deficient mice is mainly due to impairment in the fusion of cytoplasmic tubulovesicular structures to the apical canalicular plasma membrane. Among the molecules involved in the membrane fusion event, only SNAP25 showed a significant decrease in mRNA expression, which likely caused the impairment in acid secretion from parietal cells in Foxl1-deficient mice, with the reduction in H+,K(+)-ATPase expression contributing to additional effect.
Collapse
Affiliation(s)
- Yasutaka Kato
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Nguyen NV, Gleeson PA, Courtois-Coutry N, Caplan MJ, Van Driel IR. Gastric parietal cell acid secretion in mice can be regulated independently of H/K ATPase endocytosis. Gastroenterology 2004; 127:145-54. [PMID: 15236181 DOI: 10.1053/j.gastro.2004.04.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Gastric parietal cells secrete acid into the lumen of the stomach. They express a proton pump, the gastric H(+)/K(+) ATPase, the activity of which is tightly regulated. The H(+)/K(+) ATPase traffics between an intracytoplasmic compartment (tubulovesicles) in quiescent parietal cells and the apical plasma membrane in activated cells. These trafficking events are considered to contribute to the control of acid secretion by modulating access to apical K(+) and Cl(-) conductances that are required for transmembrane H(+) ion transport by the H(+)/K(+) ATPase. Here, we have determined whether the control of acid secretion in vivo requires membrane trafficking of the H(+)/K(+) ATPase. METHODS We developed mice that only express an H(+)/K(+) ATPase beta subunit in which a putative tyrosine-based endocytosis motif in the cytoplasmic tail is mutated. Location of the H(+)/K(+) ATPase and parietal cell ultrastructure and gastric acid secretion were then examined. RESULTS Parietal cells of these mice lacked a tubulovesicular compartment, and the H(+)/K(+) ATPase was resident exclusively on the apical plasma membrane. Despite the inability of the H(+)/K(+) ATPase to be endocytosed, the gastric acid secretory response to histamine or an antagonist was very similar to that of wild-type mice, indicating that control of H(+)/K(+) ATPase activity can occur independently of intracellular trafficking. CONCLUSIONS We were able to dissociate the regulation of H(+)/K(+) ATPase activity from intracellular trafficking of the protein. Thus, it is likely that direct regulation of apical K(+) and Cl(-) conductances are sufficient to control gastric acid secretion.
Collapse
Affiliation(s)
- Nhung V Nguyen
- The Russell Grimwade School of Biochemistry and Molecular Biology, The University of Melbourne, Australia
| | | | | | | | | |
Collapse
|
36
|
N/A. N/A. Shijie Huaren Xiaohua Zazhi 2004; 12:1397-1401. [DOI: 10.11569/wcjd.v12.i6.1397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
37
|
Banner B, Spicer Z, Alroy J. Expression of CD3 epsilon subunit in gastric parietal cells: a possible role in signal transduction? Pathol Res Pract 2004; 199:137-43. [PMID: 12812314 DOI: 10.1078/0344-0338-00366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
CD3 antigen, formerly thought to be specific for T lymphocytes, has been found on gastric parietal cells in animals and humans. The common anti-CD3 antibodies recognize the epsilon subunit, which has a role in signal transduction. The aim of this study was to immunostain stomach specimens from humans and different animal species for CD3 antigen to determine if CD3 antigen is conserved across species and if CD3 antigen expression is altered in humans by use of certain drugs or the presence of gastritis. Gastric biopsies from 50 humans and necropsy sections from 13 different animals were immunostained using commercial anti CD3 epsilon antibodies on an automated immunostainer. Sections of stomach from four mice lacking the gastric H+,K(+)-ATPase alpha-subunit and four control mice were similarly immunostained. CD3 epsilon antigen expression in cytoplasm and cell membranes of gastric parietal cells was graded subjectively based on the number of positive cells. CD3 epsilon antigen was found on gastric parietal cells in all but one species studied, with varying expression in membranes and cytoplasm. There was a trend toward a decreased frequency of CD3+ cells in biopsies from patients on drugs (n = 23) compared to those on no drugs (n = 27). This trend was most marked in patients on H2 receptor antagonists. There was no correlation between CD3 expression and inflammation or Helicobacter pylori (H. pylori) infection. There was loss of CD3 expression in parietal cells in mice lacking the alpha-subunit of H+,K(+)-ATPase. These findings support previous observations that CD3 antigen is present in gastric parietal cells, and suggest that it may function in signal transduction during acid secretion.
Collapse
Affiliation(s)
- Barbara Banner
- Department of Pathology, UMASS Memorial Healthcare and Medical School, Worcester, MA 016585, USA.
| | | | | |
Collapse
|
38
|
Asano S, Morii M, Takeguchi N. Molecular and Cellular Regulation of the Gastric Proton Pump. Biol Pharm Bull 2004; 27:1-12. [PMID: 14743830 DOI: 10.1248/bpb.27.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The gastric H+, K+-ATPase is a proton pump that is responsible for gastric acid secretion and that actively transports protons and K+ ions in opposite directions to generate in excess of a million-fold gradient across the membrane under physiological conditions. This pump is also a target molecule of proton pump inhibitors which are used for the clinical treatment of hyperacidity. In this review, we wish to summarize the molecular regulation of this pump based on mutational studies, particularly those used for the identification of binding sites for cations and specific inhibitors. Recent reports by Toyoshima et al (2000, 2002) presented precise three-dimensional (3-D) structures of the sarcoplasmic reticulum (SR) Ca2+-ATPase, which belongs to the same family as the gastric H+, K+-ATPase. We have studied the structure-function relationships for the gastric H+, K+-ATPase using 3-D structures constructed by homology modeling of the related SR Ca2+-ATPase, which was used as a template molecule. We also discuss in this review, the regulation of cell surface expression and synthesis control of the gastric proton pump.
Collapse
Affiliation(s)
- Shinji Asano
- Life Scientific Research Center, Toyama Medican and Pharmaceutical University, 2630 Sugitani, Toyama 930-0194, Japan.
| | | | | |
Collapse
|
39
|
Abstract
AIM: To investigate the gastrin secreting cells (G cells) and the somatostatin secreting cells (D cells) of antral mucosa in rats at the ultrastructural level.
METHODS: Revised immunoelectron microscopic technique was used to detect the G cells and D cells in rat antral mucosa through gastrin and somatostatin antibodies labeled by colloidal gold. Also the relevant quantitative analysis regarding the granular number of colloidal gold in G cells and in D cells was conducted.
RESULTS: Immunological granules of colloidal gold were distributed in G cells and D cells. Gastrin labeled golden granules or somatostatin labeled ones presented mainly as lobation-like or island-like congeries. Most of the golden congeries were observed dissociated in cytoplasms of G cells or D cells, near the basement membrane. A few golden congeries were located in nuclei. The number of golden granules in one G cell was around 107.04 ± 19.68 and was 83.36 ± 17.58 in one D cell.
CONCLUSION: Gastrin secreting granules are located in cytoplasms and nuclei of G cells, and somatostatin secreting granules both in cytoplasms and in nuclei of D cells. The number of golden granules can be quantitatively analyzed to determine the relative amount of gastrin secreting granules or somatostatin secreting granules.
Collapse
Affiliation(s)
- Feng-Peng Sun
- Department of Gastroenterology, Nanfang Hospital, First Military Medical University, Guangzhou 510515, Guangdong Province, China.
| | | |
Collapse
|
40
|
Duffield A, Kamsteeg EJ, Brown AN, Pagel P, Caplan MJ. The tetraspanin CD63 enhances the internalization of the H,K-ATPase beta-subunit. Proc Natl Acad Sci U S A 2003; 100:15560-5. [PMID: 14660791 PMCID: PMC307607 DOI: 10.1073/pnas.2536699100] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The tetraspanin CD63 resides in late endosomes, lysosomes, secretory vesicles, and at the plasma membrane, and it moves among these compartments. We find that CD63 is present also in tubulovesicular elements, the intracellular compartments that contain the H,K-ATPase in unstimulated gastric parietal cells. The H,K-ATPase beta-subunit and CD63 colocalize in parietal cells and form a complex that can be coprecipitated. The beta-subunit and CD63 also interact when they are coexpressed in COS-7 cells. Furthermore, expression with CD63 induces the redistribution of the beta-subunit from the cell surface to CD63+ intracellular compartments. Immunofluorescence and biochemical experiments reveal that this redistribution occurs by enhanced endocytosis of H,K-ATPase beta-subunit complexed with CD63. Coexpression of the beta-subunit with mutant CD63 polypeptides demonstrates that the enhanced internalization of the beta-subunit depends on the capacity of CD63 to interact with adaptor protein complexes 2 and 3. These data indicate that CD63 serves as an adaptor protein that links its interaction partners to the endocytic machinery of the cell and suggest a previously uncharacterized protein-trafficking role for the tetraspanins.
Collapse
Affiliation(s)
- Amy Duffield
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | | | | | | | | |
Collapse
|
41
|
Tashiro K, Nagao T, Kurose H, Ichijo H, Urushidani T. Role of Rho in rabbit parietal cell. J Cell Physiol 2003; 197:409-17. [PMID: 14566970 DOI: 10.1002/jcp.10370] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Rho is known as an important regulator of actin microfilament formation. We were led to study it because a dynamic rearrangement of actin filaments occurs during activation of gastric acid secretion. In order to use specific probes, the rabbit gastric gland culture system was employed and the various genes were expressed using adenovirus vector. When the constitutive active mutant of Rho (RhoAV14) was expressed, histamine- or carbachol-stimulated acid secretion monitored by (14)C-aminopyrine accumulation was inhibited. Conversely, expression of C3 toxin, the specific inhibitor of Rho, and expression of G(12/13)-specific regulator of G-protein signaling domain, the specific inhibitor of G(12/13) which is considered to be an upstream mediator of Rho, both potentiated acid secretion stimulated by the agonists. F-actin staining of parietal cell expressing RhoAV14 revealed that the microfilament supporting the intracellular canaliculi (not on the basolateral membrane) almost disappeared. No clear changes in the intracellular localization of Rho were observed during stimulation of parietal cell. In resting glands, the endogenous active form of Rho was relatively high, and it decreased during histamine stimulation. The finding that any treatment which inhibit Rho augment acid secretion whereas those that activate Rho inhibit secretion strongly suggests that the Rho-pathway conducts a negatively regulating signal in parietal cell activation, possibly via site-specific regulation of actin microfilaments.
Collapse
Affiliation(s)
- Keiichiro Tashiro
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo
| | | | | | | | | |
Collapse
|
42
|
Lapierre LA, Dorn MC, Zimmerman CF, Navarre J, Burnette JO, Goldenring JR. Rab11b resides in a vesicular compartment distinct from Rab11a in parietal cells and other epithelial cells. Exp Cell Res 2003; 290:322-31. [PMID: 14567990 DOI: 10.1016/s0014-4827(03)00340-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The Rab11 family of small GTPases is composed of three members, Rab11a, Rab11b, and Rab25. While recent work on Rab11a and Rab25 has yielded some insights into their function, Rab11b has received little attention. Therefore, we sought to examine the distribution of endogenous Rab11b in epithelial cells. In rabbit gastric parietal cells, unlike Rab11a, Rab11b did not colocalize or coisolate with H(+)/K(+)-ATPase. In MDCK cells, endogenous Rab11b localized to an apical pericentrisomal region distinct from Rab11a. The microtubule agents nocodazole and taxol dramatically alter Rab11a's localization in the cell, while effects on Rab11b's distribution were less apparent. These results indicate that in contrast to Rab11a, the Rab11b compartment in the apical region is not as dependent upon microtubules. While Rab11a is known to regulate transferrin trafficking in nonpolarized cells and IgA trafficking in polarized cells, Rab11b exhibited little colocalization with either of these cargoes. Thus, while Rab11a and Rab11b share high sequence homology, they appear to reside within distinct vesicle compartments.
Collapse
Affiliation(s)
- Lynne A Lapierre
- Department of Medicine, Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, GA 30912, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Lang KS, Mueller MM, Tanneur V, Wallisch S, Fedorenko O, Palmada M, Lang F, Bröer S, Heilig CW, Schleicher E, Weigert C. Regulation of cytosolic pH and lactic acid release in mesangial cells overexpressing GLUT1. Kidney Int 2003; 64:1338-47. [PMID: 12969152 DOI: 10.1046/j.1523-1755.2003.00213.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Anaerobic glycolysis leads to the formation of lactate and H+ and thus imposes a significant challenge on cytosolic acid/base regulation. Cytosolic acidification, on the other hand, is known to inhibit flux through glycolysis and lactate formation. To explore the interplay of cytosolic pH and glycolysis, rat mesangial cells transfected with the glucose transporter GLUT1 (GLUT1 cells) were compared with those transfected with beta-galactosidase (LacZ cells). METHODS In the presence of extracellular glucose, the glycolytic rate was one order of magnitude higher in GLUT1 cells than in LacZ cells. Cytosolic pH (pHi) was significantly higher in GLUT1 than LacZ cells, an effect abolished in the presence of Na+/H+ exchange inhibitor ethylisopropylamiloride (1 micromol/L). RESULTS Addition of 40 mmol/L lactate led to marked cytosolic acidification, which was in both cell types blunted by O-methyl-glucose (20 mmol/L) and completely abolished by 100 micromol/L phloretin and 1 mmol/L p-chloromercuribenzene-sulphonic acid (p-CMBS) and in LacZ cells only by glucose (20 mmol/L). The functional characterization points to the involvement of a lactic acid transporter from the monocarboxylate transporter (MCT) family, particularly MCT1. Reverse transcription-polymerase chain reaction (RT-PCR) indeed disclosed the expression of MCT1 and MCT2 in both GLUT1 and LacZ cells. CONCLUSION Overexpression of GLUT1 leads to cytosolic alkalinization of mesangial cells depending on functional Na+/H+ exchanger but not on Na+ independent H+ transport.
Collapse
Affiliation(s)
- Karl S Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Matsukawa J, Nakayama K, Nagao T, Ichijo H, Urushidani T. Role of ADP-ribosylation factor 6 (ARF6) in gastric acid secretion. J Biol Chem 2003; 278:36470-5. [PMID: 12860984 DOI: 10.1074/jbc.m305444200] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ADP-ribosylation factor (ARF) proteins are monomeric GTPases that are essential for membrane transport and exocytosis in a number of secretory cells. We investigated ARF6, the activation of which is insensitive to brefeldin A, to determine whether it regulates membrane traffic in gastric parietal cells. ARF6 translocated from cytosol to tubulovesicle in the presence of GTPgammaS, a potential inhibitor of acid secretion in permeabilized cells, whereas under the Mg2+-chelated condition where activity of ARF-GTPase activating protein is inhibited, ARF6 translocated to the apical secretory membrane. Immunohistochemical examination revealed that ARF6 mainly located in parietal cell within the gastric glands, and it translocated from the cytosol to the intracellular canaliculi when the glands were stimulated. These results indicated that the distribution of ARF6 between cytosol and the two different membranes was regulated by its GTPase activity. In cultured gastric glands infected with adenovirus expressing ARF6 Q67L, a mutant lacking GTP hydrolysis activity, gastric acid secretion was inhibited. These results suggest that ARF6 regulates gastric acid secretion in parietal cell and that the GTP hydrolysis cycle of ARF6 is essential for the activation pathway.
Collapse
Affiliation(s)
- Jun Matsukawa
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | | | |
Collapse
|
45
|
Machen TE, Leigh MJ, Taylor C, Kimura T, Asano S, Moore HPH. pH of TGN and recycling endosomes of H+/K+-ATPase-transfected HEK-293 cells: implications for pH regulation in the secretory pathway. Am J Physiol Cell Physiol 2003; 285:C205-14. [PMID: 12660145 DOI: 10.1152/ajpcell.00008.2003] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The influences of the gastric H+/K+ pump on organelle pH during trafficking to and from the plasma membrane were investigated using HEK-293 cells stably expressing the alpha- and beta-subunits of human H+/K+-ATPase (H+/K+-alpha,beta cells). The pH values of trans-Golgi network (pHTGN) and recycling endosomes (pHRE) were measured by transfecting H+/K+-alpha,beta cells with the pH-sensitive GFP pHluorin fused to targeting sequences of either TGN38 or synaptobrevin, respectively. Immunofluorescence showed that H+/K+-ATPase was present in the plasma membrane, TGN, and RE. The pHTGN was similar in both H+/K+-alpha,beta cells (pHTGN 6.36) and vector-transfected ("mock") cells (pHTGN 6.34); pHRE was also similar in H+/K+-alpha,beta (pHRE 6.40) and mock cells (pHRE 6.37). SCH28080 (inhibits H+/K+-ATPase) caused TGN to alkalinize by 0.12 pH units; subsequent addition of bafilomycin (inhibits H+ v-ATPase) caused TGN to alkalinize from pH 6.4 up to a new steady-state pHTGN of 7.0-7.5, close to pHcytosol. Similar results were observed in RE. Thus H+/K+-ATPases that trafficked to the plasma membrane were active but had small effects to acidify the TGN and RE compared with H+ v-ATPase. Mathematical modeling predicted a large number of H+ v-ATPases (8000) active in the TGN to balance a large, passive H+ leak (with PH approximately 10-3 cm/s) via unidentified pathways out of the TGN. We propose that in the presence of this effective, though inefficient, buffer system in the Golgi and TGN, H+/K+-ATPases (estimated to be approximately 4000 active in the TGN) and other transporters have little effect on luminal pH as they traffic to the plasma membrane.
Collapse
Affiliation(s)
- Terry E Machen
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Samuelson LC, Hinkle KL. Insights into the regulation of gastric acid secretion through analysis of genetically engineered mice. Annu Rev Physiol 2003; 65:383-400. [PMID: 12517996 DOI: 10.1146/annurev.physiol.65.092101.142213] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The regulation of acid secretion in the stomach involves a complex network of factors that stimulate secretion in response to the ingestion of a meal and maintain homeostasis of gastric pH. Genetically engineered mouse models have provided a new opportunity to investigate the importance and function of specific molecules and pathways involved in the regulation of acid secretion. Mouse mutants with disruptions in the three major stimulatory pathways for acid secretion in parietal cells, gastrin, histamine, and acetylcholine, have been generated. Disruption of the gastrin pathway results in a major impairment in both basal and induced acid secretion. Histamine and acetylcholine pathway mutants also have significant alterations in acid secretion, although the impairment does not appear to be as severe as in gastrin pathway mutants, perhaps due in part to the hypergastrinemia that occurs. Mice with a disruption in the somatostatin pathway have increased gastric acid secretion, which confirms an important negative regulatory role for this factor. This review discusses these genetically engineered mouse models, as well as others, that provide insight into the complex regulation of in vivo gastric acid secretion. The regulation of growth and cellular morphology of the stomach in these mouse models is also presented. In addition, transgene promoters that are expressed in the gastric epithelium are discussed because these promoters will be important tools to alter cellular physiology in new mouse models in the future.
Collapse
Affiliation(s)
- Linda C Samuelson
- Department of Physiology, The University of Michigan, Ann Arbor, Michigan, 48109-0622, USA.
| | | |
Collapse
|
47
|
Royle SJ, Murrell-Lagnado RD. Constitutive cycling: a general mechanism to regulate cell surface proteins. Bioessays 2003; 25:39-46. [PMID: 12508281 DOI: 10.1002/bies.10200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cells can change their function by rapidly modulating the levels of certain proteins at the plasma membrane. This rapid modulation is achieved by using a specialised trafficking process called constitutive cycling. The constitutive cycling of a variety of transmembrane proteins such as receptors, channels and transporters has recently been directly demonstrated in a wide range of cell types. This regulation is thought to underlie important biological phenomena such as learning and memory, gastric acid secretion and water and blood glucose homeostasis. This review discusses the molecular mechanisms of constitutive cycling, its regulation by extracellular agents such as hormones and its misregulation in disease states.
Collapse
Affiliation(s)
- Stephen J Royle
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK.
| | | |
Collapse
|
48
|
Karvar S, Yao X, Crothers JM, Liu Y, Forte JG. Localization and function of soluble N-ethylmaleimide-sensitive factor attachment protein-25 and vesicle-associated membrane protein-2 in functioning gastric parietal cells. J Biol Chem 2002; 277:50030-5. [PMID: 12386166 DOI: 10.1074/jbc.m207694200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The soluble N-ethylmaleimide-sensitive factor attachment protein of 25 kDa (SNAP-25) plays an important role in vesicle trafficking. Together with vesicle-associated membrane protein-2 (VAMP-2) and syntaxin, SNAP-25 forms a ternary complex implicated in docking and fusion of secretory vesicles with the plasma membrane during exocytosis. These so-called SNARE proteins are believed to regulate tubulovesicle trafficking and fusion during the secretory cycle of the gastric parietal cell. Here we examined the cellular localization and functional importance of SNAP-25 in parietal cell cultures. Adenoviral constructs were used to express SNAP-25 tagged with cyan fluorescent protein, VAMP-2 tagged with yellow fluorescent protein, and SNAP-25 in which the C-terminal 25 amino acids were deleted (SNAP-25 Delta181-206). Membrane fractionation experiments and fluorescent imaging showed that SNAP-25 is localized to the apical plasma membrane. The expression of the mutant SNAP-25 Delta181-226 inhibited the acid secretory response of parietal cells. Also, SNAP Delta181-226 bound poorly in vitro with recombinant syntaxin-1 compared with wild type SNAP-25, indicating that pairing between syntaxin-1 and SNAP-25 is required for parietal cell activation. Dual expression of SNAP-25 tagged with cyan fluorescent protein and VAMP-2 tagged with yellow fluorescent protein revealed a dynamic change in distribution associated with acid secretion. In resting cells, SNAP-25 is at the apical plasma membrane and VAMP-2 is associated with cytoplasmic H,K-ATPase-rich tubulovesicles. After stimulation, the two proteins co-localize on the apical plasma membrane. These data demonstrate the functional significance of SNAP-25 as a SNARE protein in the parietal cell and show the dynamic stimulation-associated redistribution of VAMP-2 from H,K-ATPase-rich tubulovesicles to co-localize with SNAP-25 on the apical plasma membrane.
Collapse
Affiliation(s)
- Serhan Karvar
- Department of Molecular & Cell Biology, University of California, Berkeley, CA 94720, USA
| | | | | | | | | |
Collapse
|
49
|
Matsukawa J, Tashiro K, Nagao T, Urushidani T. Role of small GTP-binding proteins and cytoskeleton in gastric acid secretion. Inflammopharmacology 2002. [DOI: 10.1163/156856002321544800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
50
|
Akhter S, Kovbasnjuk O, Li X, Cavet M, Noel J, Arpin M, Hubbard AL, Donowitz M. Na(+)/H(+) exchanger 3 is in large complexes in the center of the apical surface of proximal tubule-derived OK cells. Am J Physiol Cell Physiol 2002; 283:C927-40. [PMID: 12176749 DOI: 10.1152/ajpcell.00613.2001] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell biological approaches were used to examine the location and function of the brush border (BB) Na(+)/H(+) exchanger NHE3 in the opossum kidney (OK) polarized renal proximal tubule cell line. NHE3 epitope tagged with the vesicular stomatitis virus glycoprotein epitope (NHE3V) was stably expressed and called OK-E3V cells. On the basis of cell surface biotinylation studies, these cells had 10-15% of total NHE3 on the BB. Intracellular NHE3V largely colocalized with Rab11 and to a lesser extent with EEA1. The BB location of NHE3V was examined by confocal microscopy relative to the lectins wheat germ aggluttinin (WGA) and phytohemagluttin E (PHA-E), as well as the B subunit of cholera toxin (CTB). The cells were pyramidal, and NHE3 was located in microvilli in the center of the apical surface. In contrast, PHA-E, WGA, and CTB were diffusely distributed on the BB. Detergent extraction showed that total NHE3V was largely soluble in Triton X-100, whereas virtually all surface NHE3V was insoluble. Sucrose density gradient centrifugation demonstrated that total NHE3V migrated at the same size as approximately 400- and approximately 900-kDa standards, whereas surface NHE3V was enriched in the approximately 900-kDa form. Under basal conditions, NHE3 cycled between the cell surface and the recycling pathway through a phosphatidylinositol (PI) 3-kinase-dependent mechanism. Measurements of surface and intracellular pH were obtained by using FITC-WGA. Internalization of FITC-WGA occurred largely into the juxtanuclear compartment that contained Rab11 and NHE3V. pH values on the apical surface and in endosomes in the presence of the NHE3 blocker, S3226, were elevated, showing that NHE3 functioned to acidify both compartments. In conclusion, NHE3V in OK cells exists in distinct domains both in the center of the apical surface and in a juxtanuclear compartment. In the BB fraction, NHE3 is largely in the detergent-insoluble fraction in lipid rafts and/or in large heterogenous complexes ranging from approximately 400 to approximately 900 kDa.
Collapse
Affiliation(s)
- S Akhter
- Department of Medicine, Gastrointestinal Division, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205-2195, USA
| | | | | | | | | | | | | | | |
Collapse
|