1
|
Ljungholm PL, Ermund A, Söderlund Garsveden MM, Pettersson VL, Gustafsson JK. The anion exchanger slc26a3 regulates colonic mucus expansion during steady state and in response to prostaglandin E 2, while Cftr regulates de novo mucus release in response to carbamylcholine. Pflugers Arch 2024; 476:1209-1219. [PMID: 38829391 PMCID: PMC11271379 DOI: 10.1007/s00424-024-02975-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/05/2024]
Abstract
The intestinal epithelium is covered by mucus that protects the tissue from the luminal content. Studies have shown that anion secretion via the cystic fibrosis conductance regulator (Cftr) regulates mucus formation in the small intestine. However, mechanisms regulating mucus formation in the colon are less understood. The aim of this study was to explore the role of anion transport in the regulation of mucus formation during steady state and in response to carbamylcholine (CCh) and prostaglandin E2 (PGE2). The broad-spectrum anion transport inhibitor 4,4'-diisothiocyanatostilbene-2,2'-disulfonate (DIDS), CftrdF508 (CF) mice, and the slc26a3 inhibitor SLC26A3-IN-2 were used to inhibit anion transport. In the distal colon, steady-state mucus expansion was reduced by SLC26A3-IN-2 and normal in CF mice. PGE2 stimulated mucus expansion without de novo mucus release in wild type (WT) and CF colon via slc26a3 sensitive mechanisms, while CCh induced de novo mucus secretion in WT but not in CF colon. However, when added simultaneously, CCh and PGE2 stimulated de novo mucus secretion in the CF colon via DIDS-sensitive pathways. A similar response was observed in CF ileum that responded to CCh and PGE2 with DIDS-sensitive de novo mucus secretion. In conclusion, this study suggests that slc26a3 regulates colonic mucus expansion, while Cftr regulates CCh-induced de novo mucus secretion from ileal and distal colon crypts. Furthermore, these findings demonstrate that in the absence of a functional Cftr channel, parallel stimulation with CCh and PGE2 activates additional anion transport processes that help release mucus from intestinal goblet cells.
Collapse
Affiliation(s)
- Penny L Ljungholm
- Department of Physiology, University of Gothenburg, Medicinaregatan 11, Box 432, 405 30, Gothenburg, Sweden
| | - Anna Ermund
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | | | - Victor L Pettersson
- Department of Physiology, University of Gothenburg, Medicinaregatan 11, Box 432, 405 30, Gothenburg, Sweden
| | - Jenny K Gustafsson
- Department of Physiology, University of Gothenburg, Medicinaregatan 11, Box 432, 405 30, Gothenburg, Sweden.
- Department of Medical Chemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
2
|
Lange F, Porath K, Sellmann T, Einsle A, Jaster R, Linnebacher M, Köhling R, Kirschstein T. Direct-Current Electrical Field Stimulation of Patient-Derived Colorectal Cancer Cells. BIOLOGY 2023; 12:1032. [PMID: 37508461 PMCID: PMC10376471 DOI: 10.3390/biology12071032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Several cues for a directional migration of colorectal cancer cells were identified as being crucial in tumor progression. However, galvanotaxis, the directional migration in direct-current electrical fields, has not been investigated so far. Therefore, we asked whether direct-current electrical fields could be used to mobilize colorectal cancer cells along field vectors. For this purpose, five patient-derived low-passage cell lines were exposed to field strengths of 150-250 V/m in vitro, and migration along the field vectors was investigated. To further study the role of voltage-gated calcium channels on galvanotaxis and intracellular signaling pathways that are associated with migration of colorectal cancer cells, the cultures were exposed to selective inhibitors. In three out of five colorectal cancer cell lines, we found a preferred cathodal migration. The cellular integrity of the cells was not impaired by exposure of the cells to the selected field strengths. Galvanotaxis was sensitive to inhibition of voltage-gated calcium channels. Furthermore, signaling pathways such as AKT and MEK, but not STAT3, were also found to contribute to galvanotaxis in our in vitro model system. Overall, we identify electrical fields as an important contributor to the directional migration of colorectal cancer cells.
Collapse
Affiliation(s)
- Falko Lange
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Katrin Porath
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Tina Sellmann
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Anne Einsle
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Robert Jaster
- Division of Gastroenterology and Endocrinology, Department of Medicine II, Rostock University Medical Center, 18057 Rostock, Germany
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Rüdiger Köhling
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Timo Kirschstein
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
3
|
Logue MJE, Farquhar RE, Eckhoff-Björngard Y, Cheung TT, Devor DC, McDonald FJ, Hamilton KL. The exocyst complex is required for the trafficking and delivery of KCa3.1 to the basolateral membrane of polarized epithelia. Am J Physiol Cell Physiol 2023; 324:C1249-C1262. [PMID: 37125772 PMCID: PMC10243536 DOI: 10.1152/ajpcell.00374.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 05/02/2023]
Abstract
Control of the movement of ions and water across epithelia is essential for homeostasis. Changing the number or activity of ion channels at the plasma membrane is a significant regulator of epithelial transport. In polarized epithelia, the intermediate-conductance calcium-activated potassium channel, KCa3.1 is delivered to the basolateral membrane where it generates and maintains the electrochemical gradients required for epithelial transport. The mechanisms that control the delivery of KCa3.1 to the basolateral membrane are still emerging. Herein, we investigated the role of the highly conserved tethering complex exocyst. In epithelia, exocyst is involved in the tethering of post-Golgi secretory vesicles with the basolateral membrane, which is required before membrane fusion. In our Fisher rat thyroid cell line that stably expresses KCa3.1, siRNA knockdown of either of the exocyst subunits Sec3, Sec6, or Sec8 significantly decreased KCa3.1-specific current. In addition, knockdown of exocyst complex subunits significantly reduced the basolateral membrane protein level of KCa3.1. Finally, co-immunoprecipitation experiments suggest associations between Sec6 and KCa3.1, but not between Sec8 and KCa3.1. Collectively, based on these data and our previous studies, we suggest that components of exocyst complex are crucially important in the tethering of KCa3.1 to the basolateral membrane. After which, Soluble N-ethylmaleimide-sensitive factor (SNF) Attachment Receptors (SNARE) proteins aid in the insertion of KCa3.1-containing vesicles into the basolateral membrane of polarized epithelia.NEW & NOTEWORTHY Our Ussing chamber and immunoblot experiments demonstrate that when subunits of the exocyst complex were transiently knocked down, this significantly reduced the basolateral population and functional expression of KCa3.1. These data suggest, combined with our protein association experiments, that the exocyst complex regulates the tethering of KCa3.1-containing vesicles to the basolateral membrane prior to the SNARE-dependent insertion of channels into the basolateral membrane of epithelial cells.
Collapse
Affiliation(s)
- Matthew J E Logue
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rachel E Farquhar
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Yoakim Eckhoff-Björngard
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Tanya T Cheung
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Daniel C Devor
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Fiona J McDonald
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Kirk L Hamilton
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
4
|
Dolan B, Ermund A, Martinez-Abad B, Johansson ME, Hansson GC. Clearance of small intestinal crypts involves goblet cell mucus secretion by intracellular granule rupture and enterocyte ion transport. Sci Signal 2022; 15:eabl5848. [PMID: 36126118 PMCID: PMC9749883 DOI: 10.1126/scisignal.abl5848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Goblet cells in the small intestinal crypts contain large numbers of mucin granules that are rapidly discharged to clean bacteria from the crypt. Because acetylcholine released by neuronal and nonneuronal cells controls many aspects of intestinal epithelial function, we used tissue explants and organoids to investigate the response of the small intestinal crypt to cholinergic stimulation. The activation of muscarinic acetylcholine receptors initiated a coordinated and rapid emptying of crypt goblet cells that flushed the crypt contents into the intestinal lumen. Cholinergic stimulation induced an expansion of the granule contents followed by intracellular rupture of the mucin granules. The mucus expanded intracellularly before the rupture of the goblet cell apical membrane and continued to expand after its release into the crypt lumen. The goblet cells recovered from membrane rupture and replenished their stores of mucin granules. Mucus secretion from the goblet cells depended on Ca2+ signaling and the expansion of the mucus in the crypt depended on gap junctions and on ion and water transport by enterocytes adjacent to the goblet cells. This distinctive mode of mucus secretion, which we refer to as "expanding secretion," efficiently cleans the small intestine crypt through coordinated mucus, ion, and fluid secretion by goblet cells and enterocytes.
Collapse
Affiliation(s)
- Brendan Dolan
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Anna Ermund
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Beatriz Martinez-Abad
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Malin E.V. Johansson
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Gunnar C. Hansson
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| |
Collapse
|
5
|
Farquhar RE, Cheung TT, Logue MJE, McDonald FJ, Devor DC, Hamilton KL. Role of SNARE Proteins in the Insertion of KCa3.1 in the Plasma Membrane of a Polarized Epithelium. Front Physiol 2022; 13:905834. [PMID: 35832483 PMCID: PMC9271999 DOI: 10.3389/fphys.2022.905834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/01/2022] [Indexed: 11/29/2022] Open
Abstract
Targeting proteins to a specific membrane is crucial for proper epithelial cell function. KCa3.1, a calcium-activated, intermediate-conductance potassium channel, is targeted to the basolateral membrane (BLM) in epithelial cells. Surprisingly, the mechanism of KCa3.1 membrane targeting is poorly understood. We previously reported that targeting of KCa3.1 to the BLM of epithelial cells is Myosin-Vc-, Rab1-and Rab8-dependent. Here, we examine the role of the SNARE proteins VAMP3, SNAP-23 and syntaxin 4 (STX-4) in the targeting of KCa3.1 to the BLM of Fischer rat thyroid (FRT) epithelial cells. We carried out immunoblot, siRNA and Ussing chamber experiments on FRT cells, stably expressing KCa3.1-BLAP/Bir-A-KDEL, grown as high-resistance monolayers. siRNA-mediated knockdown of VAMP3 reduced BLM expression of KCa3.1 by 57 ± 5% (p ≤ 0.05, n = 5). Measurements of BLM-localized KCa3.1 currents, in Ussing chambers, demonstrated knockdown of VAMP3 reduced KCa3.1 current by 70 ± 4% (p ≤ 0.05, n = 5). Similarly, siRNA knockdown of SNAP-23 reduced the expression of KCa3.1 at the BLM by 56 ± 7% (p ≤ 0.01, n = 6) and reduced KCa3.1 current by 80 ± 11% (p ≤ 0.05, n = 6). Also, knockdown of STX-4 lowered the BLM expression of KCa3.1 by 54 ± 6% (p ≤ 0.05, n = 5) and reduced KCa3.1 current by 78 ± 11% (p ≤ 0.05, n = 5). Finally, co-immunoprecipitation experiments demonstrated associations between KCa3.1, VAMP3, SNAP-23 and STX-4. These data indicate that VAMP3, SNAP-23 and STX-4 are critical for the targeting KCa3.1 to BLM of polarized epithelial cells.
Collapse
Affiliation(s)
- Rachel E. Farquhar
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Tanya T. Cheung
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Matthew J. E. Logue
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Fiona J. McDonald
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Daniel C. Devor
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, United States
| | - Kirk L. Hamilton
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- *Correspondence: Kirk L. Hamilton,
| |
Collapse
|
6
|
|
7
|
Shan W, Hu Y, Ding J, Yang X, Lou J, Du Q, Liao Q, Luo L, Xu J, Xie R. Advances in Ca 2+ modulation of gastrointestinal anion secretion and its dysregulation in digestive disorders (Review). Exp Ther Med 2020; 20:8. [PMID: 32934673 PMCID: PMC7471861 DOI: 10.3892/etm.2020.9136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/22/2020] [Indexed: 11/29/2022] Open
Abstract
Intracellular calcium (Ca2+) is a critical cell signaling component in gastrointestinal (GI) physiology. Cytosolic calcium ([Ca2+]cyt), as a secondary messenger, controls GI epithelial fluid and ion transport, mucus and neuropeptide secretion, as well as synaptic transmission and motility. The key roles of Ca2+ signaling in other types of secretory cell (including those in the airways and salivary glands) are well known. However, its action in GI epithelial secretion and the underlying molecular mechanisms have remained to be fully elucidated. The present review focused on the role of [Ca2+]cyt in GI epithelial anion secretion. Ca2+ signaling regulates the activities of ion channels and transporters involved in GI epithelial ion and fluid transport, including Cl- channels, Ca2+-activated K+ channels, cystic fibrosis (CF) transmembrane conductance regulator and anion/HCO3- exchangers. Previous studies by the current researchers have focused on this field over several years, providing solid evidence that Ca2+ signaling has an important role in the regulation of GI epithelial anion secretion and uncovering underlying molecular mechanisms. The present review is largely based on previous studies by the current researchers and provides an overview of the currently known molecular mechanisms of GI epithelial anion secretion with an emphasis on Ca2+-mediated ion secretion and its dysregulation in GI disorders. In addition, previous studies by the current researchers demonstrated that different regulatory mechanisms are in place for GI epithelial HCO3- and Cl- secretion. An increased understanding of the roles of Ca2+ signaling and its targets in GI anion secretion may lead to the development of novel strategies to inhibit GI diseases, including the enhancement of fluid secretion in CF and protection of the GI mucosa in ulcer diseases.
Collapse
Affiliation(s)
- Weixi Shan
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yanxia Hu
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jianhong Ding
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Xiaoxu Yang
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jun Lou
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qian Du
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qiushi Liao
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Lihong Luo
- Department of Oncology and Geriatrics, Traditional Chinese Medicine Hospital of Chishui City, Guizhou 564700, P.R. China
| | - Jingyu Xu
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Rui Xie
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
8
|
Vega G, Guequén A, Philp AR, Gianotti A, Arzola L, Villalón M, Zegarra-Moran O, Galietta LJ, Mall MA, Flores CA. Lack of Kcnn4 improves mucociliary clearance in muco-obstructive lung disease. JCI Insight 2020; 5:140076. [PMID: 32814712 PMCID: PMC7455130 DOI: 10.1172/jci.insight.140076] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Airway mucociliary clearance (MCC) is the main mechanism of lung defense keeping airways free of infection and mucus obstruction. Airway surface liquid volume, ciliary beating, and mucus are central for proper MCC and critically regulated by sodium absorption and anion secretion. Impaired MCC is a key feature of muco-obstructive diseases. The calcium-activated potassium channel KCa.3.1, encoded by Kcnn4, participates in ion secretion, and studies showed that its activation increases Na+ absorption in airway epithelia, suggesting that KCa3.1-induced hyperpolarization was sufficient to drive Na+ absorption. However, its role in airway epithelium is not fully understood. We aimed to elucidate the role of KCa3.1 in MCC using a genetically engineered mouse. KCa3.1 inhibition reduced Na+ absorption in mouse and human airway epithelium. Furthermore, the genetic deletion of Kcnn4 enhanced cilia beating frequency and MCC ex vivo and in vivo. Kcnn4 silencing in the Scnn1b-transgenic mouse (Scnn1btg/+), a model of muco-obstructive lung disease triggered by increased epithelial Na+ absorption, improved MCC, reduced Na+ absorption, and did not change the amount of mucus but did reduce mucus adhesion, neutrophil infiltration, and emphysema. Our data support that KCa3.1 inhibition attenuated muco-obstructive disease in the Scnn1btg/+ mice. K+ channel modulation may be a therapeutic strategy to treat muco-obstructive lung diseases. Silencing the calcium-activated potassium channel KCa.3.1 improves mucociliary clearance in muco-obstructive lung disease by decreasing sodium absorption in the airways.
Collapse
Affiliation(s)
| | - Anita Guequén
- Centro de Estudios Científicos, Valdivia, Chile.,Universidad Austral de Chile, Valdivia, Chile
| | - Amber R Philp
- Centro de Estudios Científicos, Valdivia, Chile.,Universidad Austral de Chile, Valdivia, Chile
| | | | - Llilian Arzola
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel Villalón
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Luis Jv Galietta
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Marcus A Mall
- Department of Pediatric Pulmonology, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research, Berlin, Germany
| | | |
Collapse
|
9
|
Rehman S, Narayanan K, Nickerson AJ, Coon SD, Hoque KM, Sandle GI, Rajendran VM. Parallel intermediate conductance K + and Cl - channel activity mediates electroneutral K + exit across basolateral membranes in rat distal colon. Am J Physiol Gastrointest Liver Physiol 2020; 319:G142-G150. [PMID: 32567323 PMCID: PMC7500264 DOI: 10.1152/ajpgi.00011.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Transepithelial K+ absorption requires apical K+ uptake and basolateral K+ exit. In the colon, apical H+-K+-ATPase mediates cellular K+ uptake, and it has been suggested that electroneutral basolateral K+ exit reflects K+-Cl- cotransporter-1 (KCC1) operating in parallel with K+ and Cl- channels. The present study was designed to identify basolateral transporter(s) responsible for K+ exit in rat distal colon. Active K+ absorption was determined by measuring 86Rb+ (K+ surrogate) fluxes across colonic epithelia under voltage-clamp conditions. With zero Cl- in the mucosal solution, net K+ absorption was reduced by 38%, indicating that K+ absorption was partially Cl--dependent. Serosal addition of DIOA (KCC1 inhibitor) or Ba2+ (nonspecific K+ channel blocker) inhibited net K+ absorption by 21% or 61%, respectively, suggesting that both KCC1 and K+ channels contribute to basolateral K+ exit. Clotrimazole and TRAM34 (IK channel blockers) added serosally inhibited net K+ absorption, pointing to the involvement of IK channels in basolateral K+ exit. GaTx2 (CLC2 blocker) added serosally also inhibited net K+ absorption, suggesting that CLC2-mediated Cl- exit accompanies IK channel-mediated K+ exit across the basolateral membrane. Net K+ absorption was not inhibited by serosal addition of either IbTX (BK channel blocker), apamin (SK channel blocker), chromanol 293B (KV7 channel blocker), or CFTRinh172 (CFTR blocker). Immunofluorescence studies confirmed basolateral membrane colocalization of CLC2-like proteins and Na+-K+-ATPase α-subunits. We conclude that active K+ absorption in rat distal colon involves electroneutral basolateral K+ exit, which may reflect IK and CLC2 channels operating in parallel.NEW & NOTEWORTHY This study demonstrates that during active electroneutral K+ absorption in rat distal colon, K+ exit across the basolateral membrane mainly reflects intermediate conductance K+ channels operating in conjunction with chloride channel 2, with a smaller, but significant, contribution from K+-Cl- cotransporter-1 (KCC1) activity.
Collapse
Affiliation(s)
- Shabina Rehman
- 1Departments of Biochemistry West Virginia University School of Medicine, Morgantown, West Virginia
| | - Karthikeyan Narayanan
- 1Departments of Biochemistry West Virginia University School of Medicine, Morgantown, West Virginia
| | - Andrew J. Nickerson
- 1Departments of Biochemistry West Virginia University School of Medicine, Morgantown, West Virginia,2Departments of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Steven D. Coon
- 3Department of Biological Sciences, Port Peck Community College, Poplar, Montana
| | - Kazi Mirajul Hoque
- 4Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Geoffrey I. Sandle
- 5Leeds Institute for Medical Research at St. James’s, St. James’s University Hospital. Leeds, United Kingdom
| | - Vazhaikkurichi M. Rajendran
- 1Departments of Biochemistry West Virginia University School of Medicine, Morgantown, West Virginia,6Departments of Medicine, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
10
|
Catalán MA, Julio-Kalajzić F, Niemeyer MI, Cid LP, Sepúlveda FV. Short Chain Fatty Acids Effect on Chloride Channel ClC-2 as a Possible Mechanism for Lubiprostone Intestinal Action. Cells 2020; 9:cells9081781. [PMID: 32722648 PMCID: PMC7464869 DOI: 10.3390/cells9081781] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 01/22/2023] Open
Abstract
Lubiprostone, a 20-carbon synthetic fatty acid used for the treatment of constipation, is thought to act through an action on Cl− channel ClC-2. Short chain fatty acids (SCFAs) are produced and absorbed in the distal intestine. We explore whether SCFAs affect ClC-2, re-examine a possible direct effect of lubiprostone on ClC-2, and use mice deficient in ClC-2 to stringently address the hypothesis that the epithelial effect of lubiprostone targets this anion channel. Patch-clamp whole cell recordings of ClC-2 expressed in mammalian cells are used to assay SCFA and lubiprostone effects. Using chamber measurements of ion current in mice deficient in ClC-2 or CFTR channels served to analyze the target of lubiprostone in the distal intestinal epithelium. Intracellular SCFAs had a dual action on ClC-2, partially inhibiting conduction but, importantly, facilitating the voltage activation of ClC-2. Intra- or extracellular lubiprostone had no effect on ClC-2 currents. Lubiprostone elicited a secretory current across colonic epithelia that was increased in mice deficient in ClC-2, consistent with the channel’s proposed proabsorptive function, but absent from those deficient in CFTR. Whilst SCFAs might exert a physiological effect on ClC-2 as part of their known proabsorptive effect, ClC-2 plays no part in the lubiprostone intestinal effect that appears mediated by CFTR activation.
Collapse
Affiliation(s)
- Marcelo A. Catalán
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia 5110466, Chile; (F.J.-K.); (M.I.N.); (L.P.C.)
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5090000, Chile
- Correspondence: (M.A.C.); (F.V.S.); Tel.: +56-63-2221686 (M.A.C.)
| | - Francisca Julio-Kalajzić
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia 5110466, Chile; (F.J.-K.); (M.I.N.); (L.P.C.)
| | - María Isabel Niemeyer
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia 5110466, Chile; (F.J.-K.); (M.I.N.); (L.P.C.)
| | - Luis Pablo Cid
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia 5110466, Chile; (F.J.-K.); (M.I.N.); (L.P.C.)
| | - Francisco V. Sepúlveda
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia 5110466, Chile; (F.J.-K.); (M.I.N.); (L.P.C.)
- Correspondence: (M.A.C.); (F.V.S.); Tel.: +56-63-2221686 (M.A.C.)
| |
Collapse
|
11
|
Du C, Chen S, Wan H, Chen L, Li L, Guo H, Tuo B, Dong H. Different functional roles for K + channel subtypes in regulating small intestinal glucose and ion transport. Biol Open 2019; 8:bio.042200. [PMID: 31243019 PMCID: PMC6679390 DOI: 10.1242/bio.042200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Although K+ channels are important in mediating the driving force for colonic ion transport, their role in small intestinal transport is poorly understood. To investigate this, small intestinal short circuit currents (Isc ) and HCO3 - secretion were measured in mice, and intracellular pH (pHi) was measured in small intestinal epithelial SCBN cells. The expression and location of Kv subtypes were verified by RT-PCR, western blotting and immunohistochemistry. Diabetic mice were also used to investigate the role of Kv subtypes in regulating intestinal glucose absorption. We found that KV7.1 is not involved in duodenal ion transport, while KCa3.1 selectively regulates duodenal Isc and HCO3 - secretion in a Ca2+-mediated but not cAMP-mediated manner. Blockade of KCa3.1 increased the rate of HCO3 - fluxes via cystic fibrosis transmembrane conductance regulator (CFTR) channels in SCBN cells. Jejunal Isc was significantly stimulated by glucose, but markedly inhibited by 4-aminopyridine (4-AP) and tetraethylammonium (TEA). Moreover, both Kv1.1 and Kv1.3 were expressed in jejunal mucosae. Finally, 4-AP significantly attenuated weight gain of normal and diabetic mice, and both 4-AP and TEA significantly lowered blood glucose of diabetic mice. This study not only examines the contribution of various K+ channel subtypes to small intestinal epithelial ion transport and glucose absorption, but also proposes a novel concept for developing specific K+ channel blockers to reduce weight gain and lower blood glucose in diabetes mellitus.
Collapse
Affiliation(s)
- Chao Du
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.,Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Sichuan Province, Chengdu 610000, China
| | - Siyuan Chen
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Hanxing Wan
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Lihong Chen
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi 563003, China
| | - Lingyu Li
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi 563003, China
| | - Hong Guo
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi 563003, China
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China .,Department of Medicine, School of Medicine, University of California, San Diego, CA 92093, USA
| |
Collapse
|
12
|
Rajendran VM, Sandle GI. Colonic Potassium Absorption and Secretion in Health and Disease. Compr Physiol 2018; 8:1513-1536. [PMID: 30215859 PMCID: PMC9769410 DOI: 10.1002/cphy.c170030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The colon has large capacities for K+ absorption and K+ secretion, but its role in maintaining K+ homeostasis is often overlooked. For many years, passive diffusion and/or solvent drag were thought to be the primary mechanisms for K+ absorption in human and animal colon. However, it is now clear that apical H+ ,K+ -ATPase, in coordination with basolateral K+ -Cl- cotransport and/or K+ and Cl- channels operating in parallel, mediate electroneutral K+ absorption in animal colon. We now know that K+ absorption in rat colon reflects ouabain-sensitive and ouabain-insensitive apical H+ ,K+ -ATPase activities. Ouabain-insensitive and ouabain-sensitive H+ ,K+ -ATPases are localized in surface and crypt cells, respectively. Colonic H+ ,K+ -ATPase consists of α- (HKCα ) and β- (HKCβ ) subunits which, when coexpressed, exhibit ouabain-insensitive H+ ,K+ -ATPase activity in HEK293 cells, while HKCα coexpressed with the gastric β-subunit exhibits ouabain-sensitive H+ ,K+ -ATPase activity in Xenopus oocytes. Aldosterone enhances apical H+ ,K+ -ATPase activity, HKCα specific mRNA and protein expression, and K+ absorption. Active K+ secretion, on the other hand, is mediated by apical K+ channels operating in a coordinated way with the basolateral Na+ -K+ -2Cl- cotransporter. Both Ca2+ -activated intermediate conductance K+ (IK) and large conductance K+ (BK) channels are located in the apical membrane of colonic epithelia. IK channel-mediated K+ efflux provides the driving force for Cl- secretion, while BK channels mediate active (e.g., cAMP-activated) K+ secretion. BK channel expression and activity are increased in patients with end-stage renal disease and ulcerative colitis. This review summarizes the role of apical H+ ,K+ -ATPase in K+ absorption, and apical BK channel function in K+ secretion in health and disease. © 2018 American Physiological Society. Compr Physiol 8:1513-1536, 2018.
Collapse
Affiliation(s)
| | - Geoffrey I. Sandle
- Leeds Institute of Biomedical and Clinical Sciences, St James’s University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
13
|
Philp AR, Riquelme TT, Millar-Büchner P, González R, Sepúlveda FV, Cid LP, Flores CA. Kcnn4 is a modifier gene of intestinal cystic fibrosis preventing lethality in the Cftr-F508del mouse. Sci Rep 2018; 8:9320. [PMID: 29915289 PMCID: PMC6006244 DOI: 10.1038/s41598-018-27465-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/04/2018] [Indexed: 02/07/2023] Open
Abstract
Nearly 70% of cystic fibrosis (CF) patients bear the phenylalanine-508 deletion but disease severity differs greatly, and is not explained by the existence of different mutations in compound heterozygous. Studies demonstrated that genes other than CFTR relate to intestinal disease in humans and CF-mouse. Kcnn4, the gene encoding the calcium-activated potassium channel KCa3.1, important for intestinal secretion, is present in a locus linked with occurrence of intestinal CF-disease in mice and humans. We reasoned that it might be a CF-modifier gene and bred a CF-mouse with Kcnn4 silencing, finding that lethality was almost abolished. Silencing of Kcnn4 did not improve intestinal secretory functions, but rather corrected increased circulating TNF-α level and reduced intestinal mast cell increase. Given the importance of mast cells in intestinal disease additional double mutant CF-animals were tested, one lacking mast cells (C-kitW-sh/W-sh) and Stat6-/- to block IgE production. While mast cell depletion had no effect, silencing Stat6 significantly reduced lethality. Our results show that Kcnn4 is an intestinal CF modifier gene partially acting through a STAT6-dependent mechanism.
Collapse
Affiliation(s)
- Amber R Philp
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Texia T Riquelme
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
| | - Pamela Millar-Büchner
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Rodrigo González
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
| | | | - L Pablo Cid
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile
| | - Carlos A Flores
- Centro de Estudios Científicos (CECs), Arturo Prat 514, Valdivia, Chile.
| |
Collapse
|
14
|
Julio-Kalajzić F, Villanueva S, Burgos J, Ojeda M, Cid LP, Jentsch TJ, Sepúlveda FV. K 2P TASK-2 and KCNQ1-KCNE3 K + channels are major players contributing to intestinal anion and fluid secretion. J Physiol 2017; 596:393-407. [PMID: 29143340 DOI: 10.1113/jp275178] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 11/08/2017] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS K+ channels are important in intestinal epithelium as they ensure the ionic homeostasis and electrical potential of epithelial cells during anion and fluid secretion. Intestinal epithelium cAMP-activated anion secretion depends on the activity of the (also cAMP dependent) KCNQ1-KCNE3 K+ channel, but the secretory process survives after genetic inactivation of the K+ channel in the mouse. Here we use double mutant mice to investigate which alternative K+ channels come into action to compensate for the absence of KCNQ1-KCNE3 K+ channels. Our data establish that whilst Ca2+ -activated KCa 3.1 channels are not involved, K2P two-pore domain TASK-2 K+ channels are major players providing an alternative conductance to sustain the intestinal secretory process. Work with double mutant mice lacking both TASK-2 and KCNQ1-KCNE3 channels nevertheless points to yet-unidentified K+ channels that contribute to the robustness of the cAMP-activated anion secretion process. ABSTRACT Anion and fluid secretion across the intestinal epithelium, a process altered in cystic fibrosis and secretory diarrhoea, is mediated by cAMP-activated CFTR Cl- channels and requires the simultaneous activity of basolateral K+ channels to maintain cellular ionic homeostasis and membrane potential. This function is fulfilled by the cAMP-activated K+ channel formed by the association of pore-forming KCNQ1 with its obligatory KCNE3 β-subunit. Studies using mice show sizeable cAMP-activated intestinal anion secretion in the absence of either KCNQ1 or KCNE3 suggesting that an alternative K+ conductance must compensate for the loss of KCNQ1-KCNE3 activity. We used double mutant mouse and pharmacological approaches to identify such a conductance. Ca2+ -dependent anion secretion can also be supported by Ca2+ -dependent KCa 3.1 channels after independent CFTR activation, but cAMP-dependent anion secretion is not further decreased in the combined absence of KCa 3.1 and KCNQ1-KCNE3 K+ channel activity. We show that the K2P K+ channel TASK-2 is expressed in the epithelium of the small and large intestine. Tetrapentylammonium, a TASK-2 inhibitor, abolishes anion secretory current remaining in the absence of KCNQ1-KCNE3 activity. A double mutant mouse lacking both KCNQ1-KCNE3 and TASK-2 showed a much reduced cAMP-mediated anion secretion compared to that observed in the single KCNQ1-KCNE3 deficient mouse. We conclude that KCNQ1-KCNE3 and TASK-2 play major roles in the intestinal anion and fluid secretory phenotype. The persistence of an, admittedly reduced, secretory activity in the absence of these two conductances suggests that further additional K+ channel(s) as yet unidentified contribute to the robustness of the intestinal anion secretory process.
Collapse
Affiliation(s)
| | - Sandra Villanueva
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile.,Universidad Austral de Chile, Valdivia, Chile
| | - Johanna Burgos
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile.,Universidad Austral de Chile, Valdivia, Chile
| | - Margarita Ojeda
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - L Pablo Cid
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - Thomas J Jentsch
- Leibniz-Institut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | | |
Collapse
|
15
|
Lee BSL, Devor DC, Hamilton KL. Modulation of Retrograde Trafficking of KCa3.1 in a Polarized Epithelium. Front Physiol 2017; 8:489. [PMID: 28769813 PMCID: PMC5513911 DOI: 10.3389/fphys.2017.00489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/26/2017] [Indexed: 12/14/2022] Open
Abstract
In epithelia, the intermediate conductance, Ca2+-activated K+ channel (KCa3.1) is targeted to the basolateral membrane (BLM) where this channel plays numerous roles in absorption and secretion. A growing body of research suggests that the membrane resident population of KCa3.1 may be critical in clinical manifestation of diseases. In this study, we investigated the key molecular components that regulate the degradation of KCa3.1 using a Fisher rat thyroid cell line stably expressing KCa3.1. Using immunoblot, Ussing chamber, and pharmacological approaches, we demonstrated that KCa3.1 is targeted exclusively to the BLM, provided a complete time course of degradation of KCa3.1 and degradation time courses of the channel in the presence of pharmacological inhibitors of ubiquitylation and deubiquitylation to advance our understanding of the retrograde trafficking of KCa3.1. We provide a complete degradation profile of KCa3.1 and that the degradation is via an ubiquitin-dependent pathway. Inhibition of E1 ubiquitin activating enzyme by UBEI-41 crippled the ability of the cells to internalize the channel, shown by the increased BLM surface expression resulting in an increased function of the channel as measured by a DCEBIO sensitive K+ current. Additionally, the involvement of deubiquitylases and degradation by the lysosome were also confirmed by treating the cells with PR-619 or leupeptin/pepstatin, respectively; which significantly decreased the degradation rate of membrane KCa3.1. Additionally, we provided the first evidence that KCa3.1 channels were not deubiquitylated at the BLM. These data further define the retrograde trafficking of KCa3.1, and may provide an avenue for therapeutic approach for treatment of disease.
Collapse
Affiliation(s)
- Bob Shih-Liang Lee
- Department of Physiology, School of Biomedical Sciences, University of OtagoDunedin, New Zealand
| | - Daniel C Devor
- Department of Cell Biology, University of Pittsburgh School of MedicinePittsburgh, PA, United States
| | - Kirk L Hamilton
- Department of Physiology, School of Biomedical Sciences, University of OtagoDunedin, New Zealand
| |
Collapse
|
16
|
Liu L, Zhan P, Nie D, Fan L, Lin H, Gao L, Mao X. Intermediate-Conductance-Ca2-Activated K Channel IKCa1 Is Upregulated and Promotes Cell Proliferation in Cervical Cancer. Med Sci Monit Basic Res 2017; 23:45-57. [PMID: 28280257 PMCID: PMC5358865 DOI: 10.12659/msmbr.901462] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/20/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Accumulating data point to intermediate-conductance calcium-activated potassium channel (IKCa1) as a key player in controlling cell cycle progression and proliferation of human cancer cells. However, the role that IKCa1 plays in the growth of human cervical cancer cells is largely unexplored. MATERIAL AND METHODS In this study, Western blot analysis, immunohistochemical staining, and RT-PCR were first used for IKCa1protein and gene expression assays in cervical cancer tissues and HeLa cells. Then, IKCa1 channel blocker and siRNA were employed to inhibit the functionality of IKCa1 and downregulate gene expression in HeLa cells, respectively. After these treatments, we examined the level of cell proliferation by MTT method and measured IKCa1 currents by conventional whole-cell patch clamp technique. Cell apoptosis was assessed using the Annexin V-FITC/Propidium Iodide (PI) double-staining apoptosis detection kit. RESULTS We demonstrated that IKCa1 mRNA and protein are preferentially expressed in cervical cancer tissues and HeLa cells. We also showed that the IKCa1 channel blocker, clotrimazole, and IKCa1 channel siRNA can be used to suppress cervical cancer cell proliferation and decrease IKCa1 channel current. IKCa1 downregulation by specific siRNAs induced a significant increase in the proportion of apoptotic cells in HeLa cells. CONCLUSIONS IKCa1 is overexpressed in cervical cancer tissues, and IKCa1 upregulation in cervical cancer cell linea enhances cell proliferation, partly by reducing the proportion of apoptotic cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiguang Mao
- Corresponding Authors: Xiguang Mao, e-mail: ; Lanyang Gao, e-mail:
| |
Collapse
|
17
|
Villa TG, Feijoo-Siota L, Rama JLR, Sánchez-Pérez A, de Miguel-Bouzas T. Fecal Matter Implantation as a Way to Fight Diarrhea-Causing Microorganisms. NEW WEAPONS TO CONTROL BACTERIAL GROWTH 2016:315-352. [DOI: 10.1007/978-3-319-28368-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
18
|
Henríquez C, Riquelme TT, Vera D, Julio-Kalajzić F, Ehrenfeld P, Melvin JE, Figueroa CD, Sarmiento J, Flores CA. The calcium-activated potassium channel KCa3.1 plays a central role in the chemotactic response of mammalian neutrophils. Acta Physiol (Oxf) 2016; 216:132-45. [PMID: 26138196 DOI: 10.1111/apha.12548] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/13/2015] [Accepted: 06/24/2015] [Indexed: 12/27/2022]
Abstract
AIM Neutrophils are the first cells to arrive at sites of injury. Nevertheless, many inflammatory diseases are characterized by an uncontrolled infiltration and action of these cells. Cell migration depends on volume changes that are governed by ion channel activity, but potassium channels in neutrophil have not been clearly identified. We aim to test whether KCa3.1 participates in neutrophil migration and other relevant functions of the cell. METHODS Cytometer and confocal measurements to determine changes in cell volume were used. Cells isolated from human, mouse and horse were tested for KCa3.1-dependent chemotaxis. Chemokinetics, calcium handling and release of reactive oxygen species were measured to determine the role of KCa3.1 in those processes. A mouse model was used to test for neutrophil recruitment after acute lung injury in vivo. RESULTS We show for the first time that KCa3.1 is expressed in mammalian neutrophils. When the channel is inhibited by a pharmacological blocker or by genetic silencing, it profoundly affects cell volume regulation, and chemotactic and chemokinetic properties of the cells. We also demonstrated that pharmacological inhibition of KCa3.1 did not affect calcium entry or reactive oxygen species production in neutrophils. Using a mouse model of acute lung injury, we observed that Kca3.1(-/-) mice are significantly less effective at recruiting neutrophils into the site of inflammation. CONCLUSIONS These results demonstrate that KCa3.1 channels are key actors in the migration capacity of neutrophils, and its inhibition did not affect other relevant cellular functions.
Collapse
Affiliation(s)
- C. Henríquez
- Instituto de Farmacología; Facultad de Medicina Veterinaria; Universidad Austral de Chile; Valdivia Chile
| | | | - D. Vera
- Centro de Estudios Científicos (CECs); Valdivia Chile
| | - F. Julio-Kalajzić
- Centro de Estudios Científicos (CECs); Valdivia Chile
- Pontificia Universidad Católica de Valparaíso; Valparaíso Chile
| | - P. Ehrenfeld
- Institutos de Anatomía; Histología y Patología; Universidad Austral de Chile; Valdivia Chile
| | - J. E. Melvin
- Secretory Mechanisms and Dysfunction Section; National Institute of Dental and Craniofacial Research; National Institutes of Health; Bethesda MD USA
| | - C. D. Figueroa
- Institutos de Anatomía; Histología y Patología; Universidad Austral de Chile; Valdivia Chile
| | - J. Sarmiento
- Instituto de Fisiología; Facultad de Medicina; Universidad Austral de Chile; Valdivia Chile
| | - C. A. Flores
- Centro de Estudios Científicos (CECs); Valdivia Chile
| |
Collapse
|
19
|
Genetic Inhibition Of The Ubiquitin Ligase Rnf5 Attenuates Phenotypes Associated To F508del Cystic Fibrosis Mutation. Sci Rep 2015; 5:12138. [PMID: 26183966 PMCID: PMC4505316 DOI: 10.1038/srep12138] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/17/2015] [Indexed: 12/28/2022] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the CFTR chloride channel. Deletion of phenylalanine 508 (F508del), the most frequent CF mutation, impairs CFTR trafficking and gating. F508del-CFTR mistrafficking may be corrected by acting directly on mutant CFTR itself or by modulating expression/activity of CFTR-interacting proteins, that may thus represent potential drug targets. To evaluate possible candidates for F508del-CFTR rescue, we screened a siRNA library targeting known CFTR interactors. Our analysis identified RNF5 as a protein whose inhibition promoted significant F508del-CFTR rescue and displayed an additive effect with the investigational drug VX-809. Significantly, RNF5 loss in F508del-CFTR transgenic animals ameliorated intestinal malabsorption and concomitantly led to an increase in CFTR activity in intestinal epithelial cells. In addition, we found that RNF5 is differentially expressed in human bronchial epithelia from CF vs. control patients. Our results identify RNF5 as a target for therapeutic modalities to antagonize mutant CFTR proteins.
Collapse
|
20
|
Bijvelds MJC, Loos M, Bronsveld I, Hellemans A, Bongartz JP, Ver Donck L, Cox E, de Jonge HR, Schuurkes JAJ, De Maeyer JH. Inhibition of Heat-Stable Toxin-Induced Intestinal Salt and Water Secretion by a Novel Class of Guanylyl Cyclase C Inhibitors. J Infect Dis 2015; 212:1806-15. [PMID: 25999056 DOI: 10.1093/infdis/jiv300] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/13/2015] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Many enterotoxigenic Escherichia coli strains produce the heat-stable toxin, STa, which, by activation of the intestinal receptor-enzyme guanylyl cyclase (GC) C, triggers an acute, watery diarrhea. We set out to identify GCC inhibitors that may be of benefit for the treatment of infectious diarrheal disease. METHODS Compounds that inhibit STa-induced cyclic guanosine 3',5'-monophosphate (cGMP) production were selected by performing cyclase assays on cells and membranes containing GCC, or the related GCA. The effect of leads on STa/GCC-dependent activation of the cystic fibrosis transmembrane conductance regulator anion channel was investigated in T84 cells, and in porcine and human intestinal tissue. Their effect on STa-provoked fluid transport was assessed in ligated intestinal loops in piglets. RESULTS Four N-2-(propylamino)-6-phenylpyrimidin-4-one-substituted piperidines were shown to inhibit GCC-mediated cellular cGMP production. The half maximal inhibitory concentrations were ≤ 5 × 10(-7) mol/L, whereas they were >10 times higher for GCA. In T84 monolayers, these leads blocked STa/GCC-dependent, but not forskolin/adenylyl cyclase-dependent, cystic fibrosis transmembrane conductance regulator activity. GCC inhibition reduced STa-provoked anion secretion in pig jejunal tissue, and fluid retention and cGMP levels in STa-exposed loops. These GCC inhibitors blocked STa-provoked anion secretion in rectal biopsy specimens. CONCLUSIONS We have identified a novel class of GCC inhibitors that may form the basis for development of future therapeutics for (infectious) diarrheal disease.
Collapse
Affiliation(s)
- Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Michaela Loos
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Inez Bronsveld
- Department of Pulmonology, University Medical Center Utrecht, The Netherlands
| | | | | | - Luc Ver Donck
- Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
21
|
Rabjerg M, Oliván-Viguera A, Hansen LK, Jensen L, Sevelsted-Møller L, Walter S, Jensen BL, Marcussen N, Köhler R. High expression of KCa3.1 in patients with clear cell renal carcinoma predicts high metastatic risk and poor survival. PLoS One 2015; 10:e0122992. [PMID: 25848765 PMCID: PMC4388734 DOI: 10.1371/journal.pone.0122992] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 02/26/2015] [Indexed: 01/08/2023] Open
Abstract
Background Ca2+-activated K+ channels have been implicated in cancer cell growth, metastasis, and tumor angiogenesis. Here we hypothesized that high mRNA and protein expression of the intermediate-conductance Ca2+-activated K+ channel, KCa3.1, is a molecular marker of clear cell Renal Cell Carcinoma (ccRCC) and metastatic potential and survival. Methodology/Principal Findings We analyzed channel expression by qRT-PCR, immunohistochemistry, and patch-clamp in ccRCC and benign oncocytoma specimens, in primary ccRCC and oncocytoma cell lines, as well as in two ccRCC cell lines (Caki-1 and Caki-2). CcRCC specimens contained 12-fold higher mRNA levels of KCa3.1 than oncocytoma specimens. The large-conductance channel, KCa1.1, was 3-fold more highly expressed in ccRCC than in oncocytoma. KCa3.1 mRNA expression in ccRCC was 2-fold higher than in the healthy cortex of the same kidney. Disease specific survival trended towards reduction in the subgroup of high-KCa3.1-expressing tumors (p<0.08 vs. low-KCa3.1-expressing tumors). Progression-free survival (time to metastasis/recurrence) was reduced significantly in the subgroup of high-KCa3.1-expressing tumors (p<0.02, vs. low-KCa3.1-expressing tumors). Immunohistochemistry revealed high protein expression of KCa3.1 in tumor vessels of ccRCC and oncocytoma and in a subset of ccRCC cells. Oncocytoma cells were devoid of KCa3.1 protein. In a primary ccRCC cell line and Caki-1/2-ccRCC cells, we found KCa3.1-protein as well as TRAM-34-sensitive KCa3.1-currents in a subset of cells. Furthermore, Caki-1/2-ccRCC cells displayed functional Paxilline-sensitive KCa1.1 currents. Neither KCa3.1 nor KCa1.1 were found in a primary oncocytoma cell line. Yet KCa-blockers, like TRAM-34 (KCa3.1) and Paxilline (KCa1.1), had no appreciable effects on Caki-1 proliferation in-vitro. Conclusions/Significance Our study demonstrated expression of KCa3.1 in ccRCC but not in benign oncocytoma. Moreover, high KCa3.1-mRNA expression levels were indicative of low disease specific survival of ccRCC patients, short progression-free survival, and a high metastatic potential. Therefore, KCa3.1 is of prognostic value in ccRCC.
Collapse
Affiliation(s)
- Maj Rabjerg
- Department of Pathology, Odense University Hospital, DK-5000 Odense C, Denmark
- * E-mail:
| | | | - Lars Koch Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Line Jensen
- Department of Pathology, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Linda Sevelsted-Møller
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Steen Walter
- Department of Urology, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Boye L. Jensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Niels Marcussen
- Department of Pathology, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Ralf Köhler
- Aragon Institute of Health Sciences I+CS/IIS, 50009 Zaragoza, Spain
- Fundación Agencia Aragonesa para la Investigación y Desarrollo (ARAID), 50009 Zaragoza, Spain
| |
Collapse
|
22
|
Abstract
Technological advances in the large scale analysis of human genetics have generated profound insights into possible genetic contributions to chronic diseases including the inflammatory bowel diseases (IBDs), Crohn's disease and ulcerative colitis. To date, 163 distinct genetic risk loci have been associated with either Crohn's disease or ulcerative colitis, with a substantial degree of genetic overlap between these 2 conditions. Although many risk variants show a reproducible correlation with disease, individual gene associations only affect a subset of patients, and the functional contribution(s) of these risk variants to the onset of IBD is largely undetermined. Although studies in twins have demonstrated that the development of IBD is not mediated solely by genetic risk, it is nevertheless important to elucidate the functional consequences of risk variants for gene function in relevant cell types known to regulate key physiological processes that are compromised in IBD. This article will discuss IBD candidate genes that are known to be, or are suspected of being, involved in regulating the intestinal epithelial barrier and several of the physiological processes presided over by this dynamic and versatile layer of cells. This will include assembly and regulation of tight junctions, cell adhesion and polarity, mucus and glycoprotein regulation, bacterial sensing, membrane transport, epithelial differentiation, and restitution.
Collapse
|
23
|
Affiliation(s)
- Jörg D Schulzke
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany
| | - Dorothee Günzel
- Institute of Clinical Physiology, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany
| |
Collapse
|
24
|
Venglovecz V, Rakonczay Z, Gray MA, Hegyi P. Potassium channels in pancreatic duct epithelial cells: their role, function and pathophysiological relevance. Pflugers Arch 2014; 467:625-40. [PMID: 25074489 DOI: 10.1007/s00424-014-1585-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/09/2014] [Accepted: 07/18/2014] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal epithelial cells play a fundamental role in HCO3 (-) secretion, a process which is essential for maintaining the integrity of the pancreas. Although several studies have implicated impaired HCO3 (-) and fluid secretion as a triggering factor in the development of pancreatitis, the mechanism and regulation of HCO3 (-) secretion is still not completely understood. To date, most studies on the ion transporters that orchestrate ductal HCO3 (-) secretion have focussed on the role of Cl(-)/HCO3 (-) exchangers and Cl(-) channels, whereas much less is known about the role of K(+) channels. However, there is growing evidence that many types of K(+) channels are present in ductal cells where they have an essential role in establishing and maintaining the electrochemical driving force for anion secretion. For this reason, strategies that increase K(+) channel function may help to restore impaired HCO3 (-) and fluid secretion, such as in pancreatitis, and therefore provide novel directions for future pancreatic therapy. In this review, our aims are to summarize the types of K(+) channels found in pancreatic ductal cells and to discuss their individual roles in ductal HCO3 (-) secretion. We will also describe how K(+) channels are involved in pathophysiological conditions and discuss how they could act as new molecular targets for the development of therapeutic approaches to treat pancreatic diseases.
Collapse
Affiliation(s)
- Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary,
| | | | | | | |
Collapse
|
25
|
Linley J, Loganathan A, Kopanati S, Sandle GI, Hunter M. Evidence that two distinct crypt cell types secrete chloride and potassium in human colon. Gut 2014; 63:472-9. [PMID: 23740188 DOI: 10.1136/gutjnl-2013-304695] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
BACKGROUND Human colon may secrete substantial amounts of water secondary to chloride (Cl(-)) and/or potassium (K(+)) secretion in a variety of diarrhoeal diseases. Ion secretion occurs via Cl(-) and K(+) channels, which are generally assumed to be co-located in the colonocyte apical membrane, although their exact cellular sites remain unclear. OBJECTIVE To investigate the location of apical Cl(-) (CFTR) and apical K(+) (large conductance; BK) channels within human colonic epithelium. DESIGN Whole-cell patch clamp recordings were obtained from intact human colonic crypts. Specific blockers of K(+) channels and CFTR identified different types of K(+) channel and CFTR under resting conditions and after stimulating intracellular cAMP with forskolin. The BK channel β3-subunit was localised by immunostaining. RESULTS Two types of crypt cells were identified. One (73% of cells) had whole-cell currents dominated by intermediate conductance (IK) K(+) channels under resting conditions, which developed large CFTR-mediated currents in response to increasing intracellular cAMP. The other (27% of cells) had resting currents dominated by BK channels inhibited by the BK channel blocker penitrem A, but insensitive to both forskolin and the IK channel blocker clotrimazole. Immunostaining showed co-localisation of the BK channel β3-subunit and the goblet cell marker, MUC2. CONCLUSIONS In human colon, Cl(-) secretion originates from the dominant population of colonocytes expressing apical CFTR, whereas K(+) secretion is derived from a smaller population of goblet cells expressing apical BK channels. These findings provide new insights into the pathophysiology of secretory diarrhoea and should be taken into account during the development of anti-diarrhoeal drugs.
Collapse
Affiliation(s)
- John Linley
- Institute of Systems and Membrane Biology, University of Leeds, , Leeds, West Yorkshire, UK
| | | | | | | | | |
Collapse
|
26
|
Effects of Ca2+-activated potassium and inward rectifier potassium channel on the differentiation of endothelial progenitor cells from human peripheral blood. Mol Biol Rep 2014; 41:3413-23. [DOI: 10.1007/s11033-014-3203-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 01/25/2014] [Indexed: 11/26/2022]
|
27
|
Chou CC, Lunn CA, Murgolo NJ. KCa3.1: target and marker for cancer, autoimmune disorder and vascular inflammation? Expert Rev Mol Diagn 2014; 8:179-87. [DOI: 10.1586/14737159.8.2.179] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
28
|
Zhang J, Halm ST, Halm DR. Role of the BK channel (KCa1.1) during activation of electrogenic K+ secretion in guinea pig distal colon. Am J Physiol Gastrointest Liver Physiol 2012; 303:G1322-34. [PMID: 23064759 PMCID: PMC3532550 DOI: 10.1152/ajpgi.00325.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Secretagogues acting at a variety of receptor types activate electrogenic K(+) secretion in guinea pig distal colon, often accompanied by Cl(-) secretion. Distinct blockers of K(Ca)1.1 (BK, Kcnma1), iberiotoxin (IbTx), and paxilline inhibited the negative short-circuit current (I(sc)) associated with K(+) secretion. Mucosal addition of IbTx inhibited epinephrine-activated I(sc) ((epi)I(sc)) and transepithelial conductance ((epi)G(t)) consistent with K(+) secretion occurring via apical membrane K(Ca)1.1. The concentration dependence of IbTx inhibition of (epi)I(sc) yielded an IC(50) of 193 nM, with a maximal inhibition of 51%. Similarly, IbTx inhibited (epi)G(t) with an IC(50) of 220 nM and maximal inhibition of 48%. Mucosally added paxilline (10 μM) inhibited (epi)I(sc) and (epi)G(t) by ∼50%. IbTx and paxilline also inhibited I(sc) activated by mucosal ATP, supporting apical K(Ca)1.1 as a requirement for this K(+) secretagogue. Responses to IbTx and paxilline indicated that a component of K(+) secretion occurred during activation of Cl(-) secretion by prostaglandin-E(2) and cholinergic stimulation. Analysis of K(Ca)1.1α mRNA expression in distal colonic epithelial cells indicated the presence of the ZERO splice variant and three splice variants for the COOH terminus. The presence of the regulatory β-subunits K(Ca)β1 and K(Ca)β4 also was demonstrated. Immunolocalization supported the presence of K(Ca)1.1α in apical and basolateral membranes of surface and crypt cells. Together these results support a cellular mechanism for electrogenic K(+) secretion involving apical membrane K(Ca)1.1 during activation by several secretagogue types, but the observed K(+) secretion likely required the activity of additional K(+) channel types in the apical membrane.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Susan T. Halm
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Dan R. Halm
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| |
Collapse
|
29
|
Catalán MA, Flores CA, González-Begne M, Zhang Y, Sepúlveda FV, Melvin JE. Severe defects in absorptive ion transport in distal colons of mice that lack ClC-2 channels. Gastroenterology 2012; 142:346-54. [PMID: 22079595 PMCID: PMC3267842 DOI: 10.1053/j.gastro.2011.10.037] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 10/25/2011] [Accepted: 10/26/2011] [Indexed: 01/05/2023]
Abstract
BACKGROUND & AIMS The fluid secretion model predicts that intestinal obstruction disorders can be alleviated by promoting epithelial Cl(-) secretion. The adenosine 3',5'-cyclic monophosphate (cAMP)-activated anion channel CFTR mediates Cl(-)-dependent fluid secretion in the intestine. Although the role of the ClC-2 channel has not been determined in the intestine, this voltage-gated Cl(-) channel might compensate for the secretory defects observed in patients with cystic fibrosis and other chronic constipation disorders. We investigated whether mice that lack ClC-2 channels (Clcn2(-/-)) have defects in intestinal ion transport. METHODS Immunolocalization and immunoblot analyses were used to determine the cellular localization and the amount of ClC-2 expressed in mouse early distal colon (EDC) and late distal colon (LDC). Colon sheets from wild-type and Clcn2(-/-) littermates were mounted in Ussing chambers to determine transepithelial bioelectrical parameters and Na(+), K(+), and Cl(-) fluxes. RESULTS Expression of ClC-2 was higher in the basolateral membrane of surface cells in the EDC compared with the LDC, with little expression in crypts. Neither cAMP nor Ca(2+)-induced secretion of Cl(-) was affected in the EDC or LDC of Clcn2(-/-) mice, whereas the amiloride-sensitive short-circuit current was increased approximately 3-fold in Clcn2(-/-) EDC compared with control littermates. Conversely, electroneutral Na(+), K(+), and Cl(-) absorption was dramatically reduced in colons of Clcn2(-/-) mice. CONCLUSIONS Basolateral ClC-2 channels are required for colonic electroneutral absorption of NaCl and KCl. The increase in the amiloride-sensitive short-circuit current in Clcn2(-/-) mice revealed a compensatory mechanism that is activated in the colons of mice that lack the ClC-2 channel.
Collapse
Affiliation(s)
- Marcelo A. Catalán
- Secretory Mechanisms and Dysfunction Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 10 Center Drive, Building 10/Room 5N102, Bethesda, MD 20892 USA
| | | | - Mireya González-Begne
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Yan Zhang
- Secretory Mechanisms and Dysfunction Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 10 Center Drive, Building 10/Room 5N102, Bethesda, MD 20892 USA
| | | | - James E. Melvin
- Secretory Mechanisms and Dysfunction Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 10 Center Drive, Building 10/Room 5N102, Bethesda, MD 20892 USA
| |
Collapse
|
30
|
Wulff H, Castle NA. Therapeutic potential of KCa3.1 blockers: recent advances and promising trends. Expert Rev Clin Pharmacol 2012; 3:385-96. [PMID: 22111618 DOI: 10.1586/ecp.10.11] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Ca(2+)-activated K(+) channel K(Ca)3.1 regulates membrane potential and calcium signaling in erythrocytes, activated T and B cells, macrophages, microglia, vascular endothelium, epithelia, and proliferating vascular smooth muscle cells and fibroblasts. K(Ca)3.1 has therefore been suggested as a potential therapeutic target for diseases such as sickle cell anemia, asthma, coronary restenosis after angioplasty, atherosclerosis, kidney fibrosis and autoimmunity, where activation and excessive proliferation of one or more of these cell types is involved in the pathology. This article will review the physiology and pharmacology of K(Ca)3.1 and critically examine the available preclinical and clinical data validating K(Ca)3.1 as a therapeutic target.
Collapse
|
31
|
Paul G, Marchelletta RR, McCole DF, Barrett KE. Interferon-γ alters downstream signaling originating from epidermal growth factor receptor in intestinal epithelial cells: functional consequences for ion transport. J Biol Chem 2011; 287:2144-55. [PMID: 22069319 DOI: 10.1074/jbc.m111.318139] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The epidermal growth factor receptor (EGFr) regulates many cellular functions, such as proliferation, apoptosis, and ion transport. Our aim was to investigate whether long term treatment with interferon-γ (IFN-γ) modulates EGF activation of downstream signaling pathways in intestinal epithelial cells and if this contributes to dysregulation of epithelial ion transport in inflammation. Polarized monolayers of T(84) and HT29/cl.19A colonocytes were preincubated with IFN-γ prior to stimulation with EGF. Basolateral potassium transport was studied in Ussing chambers. We also studied inflamed colonic mucosae from C57BL/6 mice treated with dextran sulfate sodium or mdr1a knock-out mice and controls. IFN-γ increased intestinal epithelial EGFr expression without increasing its phosphorylation. Conversely, IFN-γ caused a significant decrease in EGF-stimulated phosphorylation of specific EGFr tyrosine residues and activation of ERK but not Akt-1. In IFNγ-pretreated cells, the inhibitory effect of EGF on carbachol-stimulated K(+) channel activity was lost. In inflamed colonic tissues, EGFr expression was significantly increased, whereas ERK phosphorylation was reduced. Thus, although it up-regulates EGFr expression, IFN-γ causes defective EGFr activation in colonic epithelial cells via reduced phosphorylation of specific EGFr tyrosine residues. This probably accounts for altered downstream signaling consequences. These observations were corroborated in the setting of colitis. IFN-γ also abrogates the ability of EGF to inhibit carbachol-stimulated basolateral K(+) currents. Our data suggest that, in the setting of inflammation, the biological effect of EGF, including the inhibitory effect of EGF on Ca(2+)-dependent ion transport, is altered, perhaps contributing to diarrheal and other symptoms in vivo.
Collapse
Affiliation(s)
- Gisela Paul
- Division of Gastroenterology, University of California, San Diego, School of Medicine, La Jolla, California 92093, USA
| | | | | | | |
Collapse
|
32
|
Bekkali N, de Jonge HR, van den Wijngaard RMJGJ, van der Steeg AFW, Bijlsma PB, Taminiau JAJM, Desjeux JF, Benninga MA. The role of rectal chloride secretion in childhood constipation. Neurogastroenterol Motil 2011; 23:1007-12. [PMID: 21848627 DOI: 10.1111/j.1365-2982.2011.01751.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Disturbance in fluid secretion, driven by chloride secretion, might play a role in constipation. However, disturbed chloride secretion in those patients has yet to be evaluated. Therefore, the aim of this study was to compare chloride secretion in rectal biopsies of children with functional constipation (FC) to those without constipation. METHODS To measure changes in short circuit current (I(sc) in μA cm(-2)) reflecting chloride secretion, intestinal biopsies from children with constipation, to either exclude or diagnose Hirschsprung's disease, and from children without constipation (controls) undergoing colonoscopy for screening of familial adenomatous polyposis, juvenile polyps or inflammatory bowel disease (IBD), were compared and studied in Ussing chambers. Following electrogenic sodium absorption blockade by amiloride, chloride secretory responses to calcium-linked (histamine, carbachol) and cAMP-linked (IBMX/forskolin) secretagogues were assessed. KEY RESULTS Ninety-six patients (46 FC) participated; nine FC patients (n = 1 congenital syndrome and n = 8 technical problems) and 13 controls (n = 6 IBD; n = 7 technical problems) were excluded. No significant difference was found in mean (±SE) basal chloride currents between children with FC and controls (9.6 ± 1.1 vs 9.2 ± 0.8; P = 0.75, respectively). Responses to calcium-linked chloride secretagogues (histamine and carbachol) were significantly higher in controls (33.0 ± 3.0 vs 24.5 ± 2.3; P = 0.03 and 33.6 ± 3.4 vs 26.4 ± 2.7; P = 0.05 following histamine and carbachol, respectively). CONCLUSIONS & INFERENCES Calcium-linked chloride secretion is disturbed in children with FC. Whether this defect occurs at the level of histamine receptors, components of receptor-linked signal transduction pathways or basolateral Ca(2+) -sensitive K(+) channels enhancing the electrical driving force for apical chloride secretion, remains to be explored.
Collapse
Affiliation(s)
- N Bekkali
- Department of Paediatric Gastroenterology and Nutrition, Emma Children's Hospital, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Hock M, Soták M, Kment M, Pácha J. The early effect of dextran sodium sulfate administration on carbachol-induced short-circuit current in distal and proximal colon during colitis development. Physiol Res 2011; 60:921-31. [PMID: 21995894 DOI: 10.33549/physiolres.932222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Increased colonic Cl(-) secretion was supposed to be a causative factor of diarrhea in inflammatory bowel diseases. Surprisingly, hyporesponsiveness to Cl(-) secretagogues was later described in inflamed colon. Our aim was to evaluate changes in secretory responses to cholinergic agonist carbachol in distal and proximal colon during colitis development, regarding secretory activity of enteric nervous system (ENS) and prostaglandins. Increased responsiveness to carbachol was observed in both distal and proximal colon after 3 days of 2 % dextran sodium sulfate (DSS) administration. It was measured in the presence of mucosal Ba(2+) to emphasize Cl(-) secretion. The described increase was abolished by combined inhibitory effect of tetrodotoxin (TTX) and indomethacin. Indomethacin also significantly reduced TTX-sensitive current. On the 7th day of colitis development responsiveness to carbachol decreased in distal colon (compared to untreated mice), but did not change in proximal colon. TTX-sensitive current did not change during colitis development, but indomethacin-sensitive current was significantly increased the 7th day. Decreased and deformed current responses to serosal Ba(2+) were observed during colitis induction, but only in proximal colon. We conclude that besides inhibitory effect of DSS on distal colon responsiveness, there is an early stimulatory effect that manifests in both distal and proximal colon.
Collapse
Affiliation(s)
- M Hock
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | | | |
Collapse
|
34
|
Roth EK, Hirtz S, Duerr J, Wenning D, Eichler I, Seydewitz HH, Amaral MD, Mall MA. The K+ channel opener 1-EBIO potentiates residual function of mutant CFTR in rectal biopsies from cystic fibrosis patients. PLoS One 2011; 6:e24445. [PMID: 21909392 PMCID: PMC3164200 DOI: 10.1371/journal.pone.0024445] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 08/10/2011] [Indexed: 12/05/2022] Open
Abstract
Background The identification of strategies to improve mutant CFTR function remains a key priority in the development of new treatments for cystic fibrosis (CF). Previous studies demonstrated that the K+ channel opener 1-ethyl-2-benzimidazolone (1-EBIO) potentiates CFTR-mediated Cl− secretion in cultured cells and mouse colon. However, the effects of 1-EBIO on wild-type and mutant CFTR function in native human colonic tissues remain unknown. Methods We studied the effects of 1-EBIO on CFTR-mediated Cl− secretion in rectal biopsies from 47 CF patients carrying a wide spectrum of CFTR mutations and 57 age-matched controls. Rectal tissues were mounted in perfused micro-Ussing chambers and the effects of 1-EBIO were compared in control tissues, CF tissues expressing residual CFTR function and CF tissues with no detectable Cl− secretion. Results Studies in control tissues demonstrate that 1-EBIO activated CFTR-mediated Cl− secretion in the absence of cAMP-mediated stimulation and potentiated cAMP-induced Cl− secretion by 39.2±6.7% (P<0.001) via activation of basolateral Ca2+-activated and clotrimazole-sensitive KCNN4 K+ channels. In CF specimens, 1-EBIO potentiated cAMP-induced Cl− secretion in tissues with residual CFTR function by 44.4±11.5% (P<0.001), but had no effect on tissues lacking CFTR-mediated Cl−conductance. Conclusions We conclude that 1-EBIO potentiates Cl−secretion in native CF tissues expressing CFTR mutants with residual Cl− channel function by activation of basolateral KCNN4 K+ channels that increase the driving force for luminal Cl− exit. This mechanism may augment effects of CFTR correctors and potentiators that increase the number and/or activity of mutant CFTR channels at the cell surface and suggests KCNN4 as a therapeutic target for CF.
Collapse
Affiliation(s)
- Eva K. Roth
- Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Department of Pediatrics III, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center, University of Heidelberg, Heidelberg, Germany
| | - Stephanie Hirtz
- Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Department of Pediatrics III, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center, University of Heidelberg, Heidelberg, Germany
| | - Julia Duerr
- Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Department of Pediatrics III, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center, University of Heidelberg, Heidelberg, Germany
| | - Daniel Wenning
- Pediatric Gastroenterology and Hepatology, Department of Pediatrics I, University of Heidelberg, Heidelberg, Germany
| | - Irmgard Eichler
- Department of Pediatrics and Adolescent Medicine, University of Vienna, Vienna, Austria
| | - Hans H. Seydewitz
- Department of Pediatrics and Adolescent Medicine, Albert Ludwigs University, Freiburg, Germany
| | - Margarida D. Amaral
- Centre of Human Genetics, National Institute of Health Dr. Ricardo Jorge, Lisboa, Portugal
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Marcus A. Mall
- Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Department of Pediatrics III, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
35
|
Schulzke JD, Andres S, Amasheh M, Fromm A, Günzel D. Anti-diarrheal mechanism of the traditional remedy Uzara via reduction of active chloride secretion. PLoS One 2011; 6:e18107. [PMID: 21479205 PMCID: PMC3068135 DOI: 10.1371/journal.pone.0018107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Accepted: 02/21/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The root extract of the African Uzara plant is used in traditional medicine as anti-diarrheal drug. It is known to act via inhibition of intestinal motility, but malabsorptive or antisecretory mechanisms are unknown yet. EXPERIMENTAL APPROACH HT-29/B6 cells and human colonic biopsies were studied in Ussing experiments in vitro. Uzara was tested on basal as well as on forskolin- or cholera toxin-induced Cl(-) secretion by measuring short-circuit current (I(SC)) and tracer fluxes of (22)Na(+) and (36)Cl(-). Para- and transcellular resistances were determined by two-path impedance spectroscopy. Enzymatic activity of the Na(+)/K(+)-ATPase and intracellular cAMP levels (ELISA) were measured. KEY RESULTS In HT-29/B6 cells, Uzara inhibited forskolin- as well as cholera toxin-induced I(SC) within 60 minutes indicating reduced active chloride secretion. Similar results were obtained in human colonic biopsies pre-stimulated with forskolin. In HT-29/B6, the effect of Uzara on the forskolin-induced I(SC) was time- and dose-dependent. Analyses of the cellular mechanisms of this Uzara effect revealed inhibition of the Na(+)/K(+)-ATPase, a decrease in forskolin-induced cAMP production and a decrease in paracellular resistance. Tracer flux experiments indicate that the dominant effect is the inhibition of the Na(+)/K(+)-ATPase. CONCLUSION AND IMPLICATIONS Uzara exerts anti-diarrheal effects via inhibition of active chloride secretion. This inhibition is mainly due to an inhibition of the Na(+)/K(+)-ATPase and to a lesser extent to a decrease in intracellular cAMP responses and paracellular resistance. The results imply that Uzara is suitable for treating acute secretory diarrhea.
Collapse
Affiliation(s)
- Jörg D Schulzke
- Department of Gastroenterology, Section of General Medicine and Nutrition, Charité, Campus Benjamin Franklin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany.
| | | | | | | | | |
Collapse
|
36
|
Föller M, Bobbala D, Koka S, Boini KM, Mahmud H, Kasinathan RS, Shumilina E, Amann K, Beranek G, Sausbier U, Ruth P, Sausbier M, Lang F, Huber SM. Functional significance of the intermediate conductance Ca2+-activated K+ channel for the short-term survival of injured erythrocytes. Pflugers Arch 2010; 460:1029-44. [PMID: 20857305 DOI: 10.1007/s00424-010-0878-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 08/19/2010] [Accepted: 08/24/2010] [Indexed: 01/30/2023]
Abstract
Increased cytosolic Ca(2+) concentrations activate Gardos K(+) channels in human erythrocytes with membrane hyperpolarization, efflux of K(+), Cl⁻, and osmotically obliged H₂O resulting in cell shrinkage, a phenomenon referred to as Gardos effect. We tested whether the Gardos effect delays colloid osmotic hemolysis of injured erythrocytes from mice lacking the Ca(2+)-activated K(+) channel K(Ca)3.1. To this end, we applied patch clamp and flow cytometry and determined in vitro as well as in vivo hemolysis. As a result, erythrocytes from K(Ca)3.1-deficient (K(Ca)3.1(-/-)) mice lacked Gardos channel activity and the Gardos effect. Blood parameters, reticulocyte count, or osmotic erythrocyte resistance, however, did not differ between K(Ca)3.1(-/-) mice and their wild-type littermates, suggesting low or absent Gardos channel activity in unstressed erythrocytes. Oxidative stress-induced Ca(2+) entry and phospholipid scrambling were significantly less pronounced in K(Ca)3.1(-/-) than in wild-type erythrocytes. Moreover, in vitro treatment with α-toxin from Staphylococcus aureus, which forms pores in the cellular membrane, resulted in significantly stronger hemolysis of K(Ca)3.1(-/-) than of wild-type erythrocytes. Intravenous injection of α-toxin induced more profound hemolysis in K(Ca)3.1(-/-) than in wild-type mice. Similarly, intra-peritoneal application of the redox-active substance phenylhydrazine, an agent for the induction of hemolytic anemia, was followed by a significantly stronger decrease of hematocrit in K(Ca)3.1(-/-) than in wild-type mice. Finally, malaria infection triggered the activation of K(Ca)3.1 and transient shrinkage of the infected erythrocytes. In conclusion, K(Ca)3.1 channel activity and Gardos effect counteract hemolysis of injured erythrocytes, thus decreasing hemoglobin release into circulating blood.
Collapse
Affiliation(s)
- Michael Föller
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Flores CA, Cid LP, Sepúlveda FV. A tale from the Crypt: splice variants of BK channels in colonic potassium secretion. J Physiol 2010; 588:1807-8. [DOI: 10.1113/jphysiol.2010.191783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
38
|
Sørensen MV, Sausbier M, Ruth P, Seidler U, Riederer B, Praetorius HA, Leipziger J. Adrenaline-induced colonic K+ secretion is mediated by KCa1.1 (BK) channels. J Physiol 2010; 588:1763-77. [PMID: 20351045 PMCID: PMC2887993 DOI: 10.1113/jphysiol.2009.181933] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 03/29/2010] [Indexed: 11/08/2022] Open
Abstract
Colonic epithelial K(+) secretion is a two-step transport process with initial K(+) uptake over the basolateral membrane followed by K(+) channel-dependent exit into the lumen. In this process the large-conductance, Ca(2+)-activated K(Ca)1.1 (BK) channel has been identified as the only apparent secretory K(+) channel in the apical membrane of the murine distal colon. The BK channel is responsible for both resting and Ca(2+)-activated colonic K(+) secretion and is up-regulated by aldosterone. Agonists (e.g. adrenaline) that elevate cAMP are potent activators of distal colonic K(+) secretion. However, the secretory K(+) channel responsible for cAMP-induced K(+) secretion remains to be defined. In this study we used the Ussing chamber to identify adrenaline-induced electrogenic K(+) secretion. We found that the adrenaline-induced electrogenic ion secretion is a compound effect dominated by anion secretion and a smaller electrically opposing K(+) secretion. Using tissue from (i) BK wildtype (BK(+/+)) and knockout (BK(/)) and (ii) cystic fibrosis transmembrane regulator (CFTR) wildtype (CFTR(+/+)) and knockout (CFTR(/)) mice we were able to isolate the adrenaline-induced K(+) secretion. We found that adrenaline-induced K(+) secretion: (1) is absent in colonic epithelia from BK(/) mice, (2) is greatly up-regulated in mice on a high K(+) diet and (3) is present as sustained positive current in colonic epithelia from CFTR(/) mice. We identified two known C-terminal BK alpha-subunit splice variants in colonic enterocytes (STREX and ZERO). Importantly, the ZERO variant known to be activated by cAMP is differentially up-regulated in enterocytes from animals on a high K(+) diet. In summary, these results strongly suggest that the adrenaline-induced distal colonic K(+) secretion is mediated by the BK channel and probably involves aldosterone-induced ZERO splice variant up-regulation.
Collapse
Affiliation(s)
- Mads V Sørensen
- Department of Physiology and Biophysics, The Water and Salt Research Center, Aarhus University, 8000 Aarhus C, Denmark
| | | | | | | | | | | | | |
Collapse
|
39
|
Balut CM, Gao Y, Luke C, Devor DC. Immunofluorescence-based assay to identify modulators of the number of plasma membrane KCa3.1 channels. Future Med Chem 2010; 2:707-13. [PMID: 20596245 PMCID: PMC2892982 DOI: 10.4155/fmc.10.182] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Intermediate conductance Ca2+-dependent K+ channels (KCa3.1) have been proposed as therapeutic targets for numerous diseases. We recently characterized the endocytic fate of these channels; leading to the possibility that this can be pharmacologically manipulated, thereby altering the number of channels (N) at the plasma membrane. RESULTS & DISCUSSION We demonstrate that plasma membrane-localized KCa3.1 can be rapidly(10 min) tagged with a fluorophore using a combination of a biotin ligase (BirA) acceptor peptide-tagged channel and an ER-localized BirA. Endocytosis of KCa3.1 was quantified using a 96-well plate format, demonstrating that the ubiquitin-activating enzyme E1 inhibitor UBEI-41, blocks the endocytosis of KCa3.1. CONCLUSION We describe a novel method for identifying modulators of KCa endocytosis and demonstrate this can be used to modulate Nat the plasma membrane. It is anticipated that altering N will provide novel therapeutic strategies for targeting these channels in disease.
Collapse
Affiliation(s)
- Corina M Balut
- Department of Cell Biology and Physiology, S331 BST, 3500 Terrace Street, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yajuan Gao
- Department of Cell Biology and Physiology, S331 BST, 3500 Terrace Street, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cliff Luke
- Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh, One Children’s Hospital Drive, 4401 Penn Avenue, Rangos, Room 7131, Pittsburgh, PA 15224, USA
| | - Daniel C Devor
- Department of Cell Biology and Physiology, S331 BST, 3500 Terrace Street, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
40
|
Yu K, Lujan R, Marmorstein A, Gabriel S, Hartzell HC. Bestrophin-2 mediates bicarbonate transport by goblet cells in mouse colon. J Clin Invest 2010; 120:1722-35. [PMID: 20407206 DOI: 10.1172/jci41129] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Accepted: 02/17/2010] [Indexed: 01/17/2023] Open
Abstract
Anion transport by the colonic mucosa maintains the hydration and pH of the colonic lumen, and its disruption causes a variety of diarrheal diseases. Cholinergic agonists raise cytosolic Ca2+ levels and stimulate anion secretion, but the mechanisms underlying this effect remain unclear. Cholinergic stimulation of anion secretion may occur via activation of Ca2+-activated Cl- channels (CaCCs) or an increase in the Cl- driving force through CFTR after activation of Ca2+-dependent K+ channels. Here we investigated the role of a candidate CaCC protein, bestrophin-2 (Best2), using Best2-/- mice. Cholinergic stimulation of anion current was greatly reduced in Best2-/- mice, consistent with our proposed role for Best2 as a CaCC. However, immunostaining revealed Best2 localized to the basolateral membrane of mucin-secreting colonic goblet cells, not the apical membrane of Cl--secreting enterocytes. In addition, in the absence of HCO3-, cholinergic-activated current was identical in control and Best2-/- tissue preparations, which suggests that most of the Best2 current was carried by HCO3-. These data delineate an alternative model of cholinergic regulation of colonic anion secretion in which goblet cells play a critical role in HCO3- homeostasis. We therefore propose that Best2 is a HCO3- channel that works in concert with a Cl:HCO3- exchanger in the apical membrane to affect transcellular HCO3- transport. Furthermore, previous models implicating CFTR in cholinergic Cl- secretion may be explained by substantial downregulation of Best2 in Cftr-/- mice.
Collapse
Affiliation(s)
- Kuai Yu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
41
|
Flores CA, Cid LP, Sepúlveda FV. Strain-dependent differences in electrogenic secretion of electrolytes across mouse colon epithelium. Exp Physiol 2010; 95:686-98. [PMID: 20154062 DOI: 10.1113/expphysiol.2009.051102] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mice have proven to be powerful models for the study of human physiology and pathophysiology. With the advent of techniques for genomic manipulation, the possibilities for studying inherited diseases in this convenient laboratory mammal are increasing by the day. It has been reported that when knocking out or otherwise modifying genes of interest in mice, the phenotype obtained can vary markedly depending on the genetic background of the animals used in the study. The aim of this work was to study whether the genetic background can influence the characteristics of fluid and electrolyte transepithelial transport in the distal colon of three mouse strains most in use in our and other laboratories. Ussing chamber recordings revealed that the colons of C57Bl/6J, Sv 129 and Black Swiss animals have distinctive responses to the calcium agonists carbachol and histamine that are not explained by the presence of different types of muscarinic and histaminergic receptors in these tissues. We have also found differences in the cAMP-activated, KCNMA1-channel-dependent potassium secretion between the strains. We interpret this to indicate a unique distribution of KCNMA1 channels in lower parts of the crypt of Sv 129 colonic epithelium compared with that of C57Bl/6J and Black Swiss animals. The reported differences should be taken into account when choosing the genetic background of animals to be used for genetic modification.
Collapse
Affiliation(s)
- Carlos A Flores
- Centro de Estudios Científicos (CECS), Avenida Arturo Prat 514, Valdivia 5110466, Chile
| | | | | |
Collapse
|
42
|
Colonic potassium handling. Pflugers Arch 2010; 459:645-56. [PMID: 20143237 DOI: 10.1007/s00424-009-0781-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 12/23/2009] [Accepted: 12/25/2009] [Indexed: 10/19/2022]
Abstract
Homeostatic control of plasma K+ is a necessary physiological function. The daily dietary K+ intake of approximately 100 mmol is excreted predominantly by the distal tubules of the kidney. About 10% of the ingested K+ is excreted via the intestine. K+ handling in both organs is specifically regulated by hormones and adapts readily to changes in dietary K+ intake, aldosterone and multiple local paracrine agonists. In chronic renal insufficiency, colonic K+ secretion is greatly enhanced and becomes an important accessory K+ excretory pathway. During severe diarrheal diseases of different causes, intestinal K+ losses caused by activated ion secretion may become life threatening. This topical review provides an update of the molecular mechanisms and the regulation of mammalian colonic K+ absorption and secretion. It is motivated by recent results, which have identified the K+ secretory ion channel in the apical membrane of distal colonic enterocytes. The directed focus therefore covers the role of the apical Ca2+ and cAMP-activated BK channel (KCa1.1) as the apparently only secretory K+ channel in the distal colon.
Collapse
|
43
|
Preston P, Wartosch L, Günzel D, Fromm M, Kongsuphol P, Ousingsawat J, Kunzelmann K, Barhanin J, Warth R, Jentsch TJ. Disruption of the K+ channel beta-subunit KCNE3 reveals an important role in intestinal and tracheal Cl- transport. J Biol Chem 2010; 285:7165-75. [PMID: 20051516 DOI: 10.1074/jbc.m109.047829] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The KCNE3 beta-subunit constitutively opens outwardly rectifying KCNQ1 (Kv7.1) K(+) channels by abolishing their voltage-dependent gating. The resulting KCNQ1/KCNE3 heteromers display enhanced sensitivity to K(+) channel inhibitors like chromanol 293B. KCNE3 was also suggested to modify biophysical properties of several other K(+) channels, and a mutation in KCNE3 was proposed to underlie forms of human periodic paralysis. To investigate physiological roles of KCNE3, we now disrupted its gene in mice. kcne3(-/-) mice were viable and fertile and displayed neither periodic paralysis nor other obvious skeletal muscle abnormalities. KCNQ1/KCNE3 heteromers are present in basolateral membranes of intestinal and tracheal epithelial cells where they might facilitate transepithelial Cl(-) secretion through basolateral recycling of K(+) ions and by increasing the electrochemical driving force for apical Cl(-) exit. Indeed, cAMP-stimulated electrogenic Cl(-) secretion across tracheal and intestinal epithelia was drastically reduced in kcne3(-/-) mice. Because the abundance and subcellular localization of KCNQ1 was unchanged in kcne3(-/-) mice, the modification of biophysical properties of KCNQ1 by KCNE3 is essential for its role in intestinal and tracheal transport. Further, these results suggest KCNE3 as a potential modifier gene in cystic fibrosis.
Collapse
Affiliation(s)
- Patricia Preston
- Leibniz-Institut für Molekulare Pharmakologie and Max-Delbrück-Centrum für Molekulare Medizin, 13125 Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Donnellan F, Keating N, Geoghegan P, Murray FE, Harvey BJP, Keely SJ. JNK mitogen-activated protein kinase limits calcium-dependent chloride secretion across colonic epithelial cells. Am J Physiol Gastrointest Liver Physiol 2010; 298:G37-44. [PMID: 19875701 DOI: 10.1152/ajpgi.00202.2009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neuroimmune agonists induce epithelial Cl(-) secretion through elevations in intracellular Ca2+ or cAMP. Previously, we demonstrated that epidermal growth factor receptor (EGFR) transactivation and subsequent ERK MAPK activation limits secretory responses to Ca2+-dependent, but not cAMP-dependent, agonists. Although JNK MAPKs are also expressed in epithelial cells, their role in regulating transport function is unknown. Here, we investigated the potential role for JNK in regulating Cl(-) secretion in T(84) colonic epithelial cells. Western blot analysis revealed that a prototypical Ca2+-dependent secretagogue, carbachol (CCh; 100 microM), induced phosphorylation of both the 46-kDa and 54-kDa isoforms of JNK. This effect was mimicked by thapsigargin (TG), which specifically elevates intracellular Ca2+, but not by forskolin (FSK; 10 microM), which elevates cAMP. CCh-induced JNK phosphorylation was attenuated by the EGFR inhibitor, tyrphostin-AG1478 (1 microM). Pretreatment of voltage-clamped T(84) cells with SP600125 (2 microM), a specific JNK inhibitor, potentiated secretory responses to both CCh and TG but not to FSK. The effects of SP600125 on CCh-induced secretion were not additive with those of the ERK inhibitor, PD98059. Finally, in apically permeabilized T(84) cell monolayers, SP600125 potentiated CCh-induced K+ conductances but not Na+/K+ATPase activity. These data demonstrate a novel role for JNK MAPK in regulating Ca2+ but not cAMP-dependent epithelial Cl(-) secretion. JNK activation is mediated by EGFR transactivation and exerts its antisecretory effects through inhibition of basolateral K+ channels. These data further our understanding of mechanisms regulating epithelial secretion and underscore the potential for exploitation of MAPK-dependent signaling in treatment of intestinal transport disorders.
Collapse
Affiliation(s)
- Fergal Donnellan
- Dept. of Molecular Medicine, Royal College of Surgeons in Ireland, RCSI Education and Research Ctr., Smurfit Bldg., Beaumont Hospital, Dublin 9, Ireland
| | | | | | | | | | | |
Collapse
|
45
|
An investigation of the occurrence and properties of the mitochondrial intermediate-conductance Ca2+-activated K+ channel mtKCa3.1. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1797:1260-7. [PMID: 20036632 DOI: 10.1016/j.bbabio.2009.12.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 12/15/2009] [Accepted: 12/19/2009] [Indexed: 12/24/2022]
Abstract
The mitochondrial intermediate-conductance Ca2+-activated K+ channel mtKCa3.1 has recently been discovered in the HCT116 colon tumor-derived cell line, which expresses relatively high levels of this protein also in the plasma membrane. Electrophysiological recordings revealed that the channel can exhibit different conductance states and kinetic modes, which we tentatively ascribe to post-translational modifications. To verify whether the localization of this channel in mitochondria might be a peculiarity of these cells or a more widespread feature we have checked for the presence of mtKCa3.1 in a few other cell lines using biochemical and electrophysiological approaches. It turned out to be present at least in some of the cells investigated. Functional assays explored the possibility that mtKCa3.1 might be involved in cell proliferation or play a role similar to that of the Shaker-type KV1.3 channel in lymphocytes, which interacts with outer mitochondrial membrane-inserted Bax thereby promoting apoptosis (Szabò, I. et al., Proc. Natl. Acad Sci. USA 105 (2008) 14861-14866). A specific KCa3.1 inhibitor however did not have any detectable effect on cell proliferation or death.
Collapse
|
46
|
Flores CA, Cid LP, Sepúlveda FV, Niemeyer MI. TMEM16 proteins: the long awaited calcium-activated chloride channels? ACTA ACUST UNITED AC 2009; 42:993-1001. [PMID: 19784506 DOI: 10.1590/s0100-879x2009005000028] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 08/12/2009] [Indexed: 11/22/2022]
Abstract
Currents mediated by calcium-activated chloride channels (CaCCs), observed for the first time in Xenopus oocytes, have been recorded in many cells and tissues ranging from different types of neurons to epithelial and muscle cells. CaCCs play a role in the regulation of excitability in neurons including sensory receptors. In addition, they are crucial mediators of chloride movements in epithelial cells where their activity regulates electrolyte and fluid transport. The roles of CaCCs, particularly in epithelia, are briefly reviewed with emphasis on their function in secretory epithelia. The recent identification by three independent groups, using different strategies, of TMEM16A as the molecular counterpart of the CaCC is discussed. TMEM16A is part of a family that has 10 other members in mice. The discovery of the potential TMEM16 anion channel activity opens the way for the molecular investigation of the role of these anion channels in specific cells and in organ physiology and pathophysiology. The identification of TMEM16A protein as a CaCC chloride channel molecule represents a great triumph of scientific perseverance and ingenuity. The varied approaches used by the three independent research groups also augur well for the solidity of the discovery.
Collapse
Affiliation(s)
- C A Flores
- Centro de Estudios Científicos (CECS), Valdivia, Chile
| | | | | | | |
Collapse
|
47
|
Perry MD, Sandle GI. Regulation of colonic apical potassium (BK) channels by cAMP and somatostatin. Am J Physiol Gastrointest Liver Physiol 2009; 297:G159-67. [PMID: 19407217 PMCID: PMC2711756 DOI: 10.1152/ajpgi.00132.2009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
High-conductance apical K+ (BK) channels are present in surface colonocytes of mammalian (including human) colon. Their location makes them well fitted to contribute to the excessive intestinal K(+) losses often associated with infective diarrhea. Since many channel proteins are regulated by phosphorylation, we evaluated the roles of protein kinase A (PKA) and phosphatases in the modulation of apical BK channel activity in surface colonocytes from rat distal colon using patch-clamp techniques, having first increased channel abundance by chronic dietary K+ enrichment. We found that PKA activation using 50 micromol/l forskolin and 5 mmol/l 3-isobutyl-1-methylxanthine stimulated BK channels in cell-attached patches and the catalytic subunit of PKA (200 U/ml) had a similar effect in excised inside-out patches. The antidiarrheal peptide somatostatin (SOM; 2 micromol/l) had a G protein-dependent inhibitory effect on BK channels in cell-attached patches, which was unaffected by pretreatment with 10 micromol/l okadaic acid (an inhibitor of protein phosphatase type 1 and type 2A) but completely prevented by pretreatment with 100 micromol/l Na+ orthovanadate and 10 micromol/l BpV (inhibitors of phosphoprotein tyrosine phosphatase). SOM also inhibited apical BK channels in surface colonocytes in human distal colon. We conclude that cAMP-dependent PKA activates apical BK channels and may enhance colonic K+ losses in some cases of secretory diarrhea. SOM inhibits apical BK channels through a phosphoprotein tyrosine phosphatase-dependent mechanism, which could form the basis of new antidiarrheal strategies.
Collapse
Affiliation(s)
- M. D. Perry
- Institute of Molecular Medicine, St James's University Hospital, Leeds, United Kingdom
| | - G. I. Sandle
- Institute of Molecular Medicine, St James's University Hospital, Leeds, United Kingdom
| |
Collapse
|
48
|
Bajwa PJ, Lee JW, Straus DS, Lytle C. Activation of PPARgamma by rosiglitazone attenuates intestinal Cl- secretion. Am J Physiol Gastrointest Liver Physiol 2009; 297:G82-9. [PMID: 19443733 DOI: 10.1152/ajpgi.90640.2008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The thiazolidinedione (TZD) drugs rosiglitazone (Ro) and pioglitazone (Po) are PPARgamma agonists in widespread clinical use as insulin-sensitizing agents in Type 2 diabetes. On the basis of recent evidence implicating PPARgamma as a positive modulator of intestinal epithelial differentiation, we hypothesized that TZD drugs might attenuate intestinal secretory function. To evaluate this possibility, we examined the effects of Ro and Po on electrogenic Cl- secretion [short-circuit current (I(sc))] in mouse intestinal segments and in cultured human intestinal epithelial cells (HT29-Cl.19A). As hypothesized, oral administration of Ro (20 mg.kg(-1).day(-1)) to mice for 8 days markedly reduced intestinal I(sc) responses to cAMP (forskolin)- and Ca2+ (carbachol)-dependent stimuli. In these Ro-treated mice, cholera toxin-induced intestinal fluid accumulation was reduced 65%. With continued Ro treatment, the I(sc) response to carbachol recovered significantly, whereas that to forskolin remained attenuated. Treatment of HT29 cells for 5 days with 10 muM Ro or Po in vitro brought about a similar hyposecretory state. In HT29 cells, the loss of cAMP-dependent Cl- secretion was attributable to a reduced expression of CFTR Cl- channel, KCNQ1 K+ channel, and Na-K-2Cl cotransporter-1 proteins. The transient loss of Ca2+-dependent Cl- secretion involved an impairment of basolateral Ca2+-stimulated K+ channel activity without a detectable loss of K(Ca)3.1 channel protein. Our results establish TZD drugs as important modulators of intestinal Cl- secretory function.
Collapse
Affiliation(s)
- Poonam J Bajwa
- Division of Biomedical Sciences, University of California, Riverside, CA 92521-0121, USA
| | | | | | | |
Collapse
|
49
|
De Marchi U, Sassi N, Fioretti B, Catacuzzeno L, Cereghetti GM, Szabò I, Zoratti M. Intermediate conductance Ca2+-activated potassium channel (KCa3.1) in the inner mitochondrial membrane of human colon cancer cells. Cell Calcium 2009; 45:509-16. [PMID: 19406468 DOI: 10.1016/j.ceca.2009.03.014] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 03/13/2009] [Accepted: 03/23/2009] [Indexed: 11/28/2022]
Abstract
Patch-clamping mitoplasts isolated from human colon carcinoma 116 cells has allowed the identification and characterization of the intermediate conductance Ca(2+)-activated K(+)-selective channel K(Ca)3.1, previously studied only in the plasma membrane of various cell types. Its identity has been established by its biophysical and pharmacological properties. Its localisation in the inner membrane of mitochondria is indicated by Western blots of subcellular fractions, by recording of its activity in mitochondria made fluorescent by a mitochondria-targeted fluorescent protein and by the co-presence of channels considered to be markers of the inner membrane. Moderate increases of mitochondrial matrix [Ca(2+)] will cause mtK(Ca)3.1 opening, thus linking inner membrane K(+) permeability and transmembrane potential to Ca(2+) signalling.
Collapse
Affiliation(s)
- Umberto De Marchi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
50
|
Hirota CL, McKay DM. Loss of Ca-mediated ion transport during colitis correlates with reduced ion transport responses to a Ca-activated K channel opener. Br J Pharmacol 2009; 156:1085-97. [PMID: 19298254 DOI: 10.1111/j.1476-5381.2009.00122.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Epithelial surface hydration is critical for proper gut function. However, colonic tissues from individuals with inflammatory bowel disease or animals with colitis are hyporesponsive to Cl(-) secretagogues. The Cl(-) secretory responses to the muscarinic receptor agonist bethanechol are virtually absent in colons of mice with dextran sodium sulphate (DSS)-induced colitis. Our aim was to define the mechanism underlying this cholinergic hyporesponsiveness. EXPERIMENTAL APPROACH Colitis was induced by 4% DSS water, given orally. Epithelial ion transport was measured in Ussing chambers. Colonic crypts were isolated and processed for mRNA expression via RT-PCR and protein expression via immunoblotting and immunolocalization. KEY RESULTS Expression of muscarinic M(3) receptors in colonic epithelium was not decreased during colitis. Short-circuit current (I(SC)) responses to other Ca(2+)-dependent secretagogues (histamine, thapsigargin, cyclopiazonic acid and calcium ionophore) were either absent or severely attenuated in colonic tissue from DSS-treated mice. mRNA levels of several ion transport molecules (a Ca(2+)-regulated Cl(-) channel, the intermediate-conductance Ca(2+)-activated K(+) channel, the cystic fibrosis transmembrane conductance regulator, the Na(+)/K(+)-ATPase pump or the Na(+)/K(+)/2Cl(-) co-transporter) were not reduced in colonic crypts from DSS-treated mice. However, protein expression of Na(+)/K(+)-ATPase alpha1 subunits was decreased twofold during colitis. Activation of Ca(2+)-activated K(+) channels increased I(SC) significantly less in DSS colons compared with control, as did the protein kinase C activator, phorbol 12-myristate 13-acetate. CONCLUSIONS AND IMPLICATIONS Decreased Na(+)/K(+)-ATPase expression probably contributes to overall epithelial hyporesponsiveness during colitis, while dysfunctional K(+) channels may account, at least partially, for lack of epithelial secretory responses to Ca(2+)-mediated secretagogues.
Collapse
Affiliation(s)
- Christina L Hirota
- Intestinal Disease Research Programme, Department of Pathology and Molecular Medicine, McMaster University, 3330 Hospital Drive Northwest, Calgary, Alberta, Canada.
| | | |
Collapse
|