1
|
Gryn-Rynko A, Hołyńska-Iwan I, Janiak MA, Olszewska-Słonina D, Amarowicz R, Graczyk R. The impact of Morus alba L. leaf extract on intestinal ion transport. An in vitro study. Biomed Pharmacother 2022; 150:112939. [PMID: 35461088 DOI: 10.1016/j.biopha.2022.112939] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/24/2022] [Accepted: 04/06/2022] [Indexed: 11/02/2022] Open
Abstract
Morus alba L. (White mulberry), is an important and popular herbal plant of the Moraceae family. It has been widely used due to its therapeutic properties, which include antidiabetic, antibacterial, anti-inflammatory, cardiovascular, and hypolpidemic activity. The present study evaluates the effects of aqueous white mulberry leaf extract on the transepithelial ion pathway in the rabbit colon epithelium (n = 48), using electrophysiological methods. In addition, the antioxidant potential and the chemical composition of the extract were determined. A mechanical-chemical stimulation with white mulberry in RH fluid (MB-RH) caused a statistically significant (p < 0.001) increase in the transepithelial electrical potential difference, from - 0.130 to - 0.685 mV. Gentle washing of the intestine with white mulberry in bumetanide, used as inhibitor of transepithelial chloride pathways, resulted in 14.8% shorter reaction than during MB-RH stimulation. There were no statistically significant differences between the electric potential values measured during stimulation with amiloride solution, used as inhibitor of transepithelial sodium pathways, and white mulberry in amilorid solution (p = 0.485). A short-term application of extract to the colon epithelium is responsible for local and reversible inhibition of chloride ion channels. The extract enhances sodium ion absorption and consequently changes the electrical potential. The effect of white mulberry extract on sodium ion transport may be related to the mechanism of hypoglycaemic activity of mulberry leaves.
Collapse
Affiliation(s)
- Anna Gryn-Rynko
- Department of Toxicology and Bromatology, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland.
| | - Iga Hołyńska-Iwan
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Michał Adam Janiak
- Department of Chemical and Physical Properties of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Dorota Olszewska-Słonina
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Ryszard Amarowicz
- Department of Chemical and Physical Properties of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Radomir Graczyk
- Department of Biology and Animal Environment, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Poland
| |
Collapse
|
2
|
Abstract
Since prehistory, human species have depended on plants for both food and medicine. Even in countries with ready access to modern medicines, alternative treatments are still highly regarded and commonly used. Unlike modern pharmaceuticals, many botanical medicines are in widespread use despite a lack of safety and efficacy data derived from controlled clinical trials and often unclear mechanisms of action. Contributing to this are the complex and undefined composition and likely multifactorial mechanisms of action and multiple targets of many botanical medicines. Here, we review the newfound importance of the ubiquitous KCNQ subfamily of voltage-gated potassium channels as targets for botanical medicines, including basil, capers, cilantro, lavender, fennel, chamomile, ginger, and Camellia, Sophora, and Mallotus species. We discuss the implications for the traditional use of these plants for disorders such as seizures, hypertension, and diabetes and the molecular mechanisms of plant secondary metabolite effects on KCNQ channels.
Collapse
Affiliation(s)
- Kaitlyn E Redford
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California 92697, USA;
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California 92697, USA;
| |
Collapse
|
3
|
Cui Y, Hou Y, Zhang H, Liu Y, Mao K, Nie H, Ding Y. Regulation of Electrolyte Permeability by Herbal Monomers in Edematous Disorders. Curr Pharm Des 2021; 27:833-839. [PMID: 32940173 DOI: 10.2174/1381612826666200917144655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/10/2020] [Indexed: 11/22/2022]
Abstract
Edema is a gradual accumulation of fluid in the interstitial tissues or luminal cavities, which is regulated by ion transport pathways and reflects dysfunction of fluid and salt homeostasis. Increasing evidence suggests that some herbal monomers significantly reduce organ/tissue edema. In this review, we briefly summarized the electrolyte permeability involved in pathomechanisms of organ edema, and the benefits of herbal monomers on ionic transport machinery, including Na+-K+-ATPase, Na+ and Cl- channels, Na+-K+-2Cl- co-transporter, etc. Pharmaceutical relevance is implicated in developing advanced strategies to mitigate edematous disorders. In conclusion, the natural herbal monomers regulate electrolyte permeability in many edematous disorders, and further basic and clinical studies are needed.
Collapse
Affiliation(s)
- Yong Cui
- Department of Anesthesiology, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Honglei Zhang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yanhong Liu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Kejun Mao
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
4
|
Chen H, Chen Y, Yang J, Wu P, Wang X, Huang C. Effect of Ginkgo biloba extract on pacemaker channels encoded by HCN gene. Herz 2020; 46:255-261. [PMID: 32435840 DOI: 10.1007/s00059-020-04933-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/19/2020] [Accepted: 04/25/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND In the present study, the electropharmacological activity of traditional Chinese medicine, Ginkgo biloba extract (GBE), on human hyperpolarization-activated nucleotide-gated (HCN) channels and the underlying "funny" currents was investigated. METHODS Standard two-electrode voltage-clamp recordings were employed to examine the properties of cloned HCN subunit currents expressed in Xenopus oocytes under controlled conditions and GBE administration. RESULTS We found that GBE irreversibly inhibited the HCN2 and HCN4 channel currents in a concentration-dependent fashion and that the HCN4 current was more sensitive to GBE compared with HCN2. In addition, GBE inhibition of the current amplitudes of HCN2 and HCN4 currents was accompanied by a decrease in the activation and deactivation kinetics. CONCLUSION The results of this study contribute toward illustrating the antiarrhythmic mechanism of GBE, which might be useful for the treatment of arrhythmia.
Collapse
Affiliation(s)
- Hui Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, 430060, Wuhan, Hubei Province, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology, 430060, Wuhan, China
| | - Yongjun Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, 430060, Wuhan, Hubei Province, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology, 430060, Wuhan, China
| | - Jing Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, 430060, Wuhan, Hubei Province, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology, 430060, Wuhan, China
| | - Pan Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, 430060, Wuhan, Hubei Province, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology, 430060, Wuhan, China
| | - Xin Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, 430060, Wuhan, Hubei Province, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology, 430060, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, 430060, Wuhan, Hubei Province, China.
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China.
- Hubei Key Laboratory of Cardiology, 430060, Wuhan, China.
| |
Collapse
|
5
|
Pang B, Xu X, Lu Y, Jin H, Yang R, Jiang C, Shao D, Liu Y, Shi J. Prediction of new targets and mechanisms for quercetin in the treatment of pancreatic cancer, colon cancer, and rectal cancer. Food Funct 2019; 10:5339-5349. [PMID: 31393490 DOI: 10.1039/c9fo01168d] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Quercetin has been widely found to exhibit anticancer activity with low toxicity and prevalence in foods. Quercetin has been reported to inhibit digestive system cancers including pancreatic cancer (PAAD) and colon cancer (COAD), but rectal cancer (READ) has not been reported. The reported mechanisms and targets are divergent. In this study, new targets and mechanisms were predicted for the influence of quercetin on PAAD, COAD, and READ using bioinformatics methods. The results showed that quercetin may target CD36 and reduce the death rate caused by PAAD by enhancing the cell adhesion, mediating the uptake of fatty acids (FAs), regulating thrombospondin-1, and stimulating the immune response. Quercetin may lower the death rate from READ by targeting SLCO1B1 and producing enhanced effects from use of this compound, inhibiting cell growth, and inducing apoptosis in tumor cells. ACADS, ALDH3B2, UGT2A3, AMH, CDKN2A, FOSL1, CD36, CFL2, CYP3A4, and MAF were identified as targets for quercetin to reduce the death rate caused by COAD. Glutathione metabolism was mainly involved in the effect of quercetin on COAD, including the enhancement of the oxidation of fatty acids, the metabolism of anticancer medications, and the stiffness of cells, and the reduction of chemical carcinogenesis, the level of anti-Müllerian hormone, the proliferation of cancer cells and transcriptional misregulation, and mediation of the activity of glutathione transferases. The combined analyses of three databases can be referred to and used to seek medications and targets that can be applied to other diseases.
Collapse
Affiliation(s)
- Bing Pang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Xiaoguang Xu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Yao Lu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Han Jin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Rongrong Yang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Chunmei Jiang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Dongyan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Yanlin Liu
- College of Enology, Northwest A&F University, 28 Xinong Road, Yangling, Shaanxi Province 712100, China
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| |
Collapse
|
6
|
Yu B, Jiang Y, Jin L, Ma T, Yang H. Role of Quercetin in Modulating Chloride Transport in the Intestine. Front Physiol 2016; 7:549. [PMID: 27932986 PMCID: PMC5120089 DOI: 10.3389/fphys.2016.00549] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/01/2016] [Indexed: 12/17/2022] Open
Abstract
Epithelial chloride channels provide the pathways for fluid secretion in the intestine. Cystic fibrosis transmembrane conductance regulator (CFTR) and calcium-activated chloride channels (CaCCs) are the main chloride channels in the luminal membrane of enterocytes. These transmembrane proteins play important roles in many physiological processes. In this study, we have identified a flavonoid quercetin as a modulator of CaCC chloride channel activity. Fluorescence quenching assay showed that quercetin activated Cl− transport in a dose-dependent manner, with EC50 ~37 μM. Short-circuit current analysis confirmed that quercetin activated CaCC-mediated Cl− currents in HT-29 cells that can be abolished by CaCCinh-A01. Ex vivo studies indicated that application of quercetin to mouse ileum and colon on serosal side resulted in activation of CFTR and CaCC-mediated Cl− currents. Notably, we found that quercetin exhibited inhibitory effect against ANO1 chloride channel activity in ANO1-expressing FRT cells and decreased mouse intestinal motility. Quercetin-stimulated short-circuit currents in mouse ileum was multi-component, which included elevation of Ca2+ concentration through L-type calcium channel and activation of basolateral NKCC, Na+/K+-ATPase, and K+ channels. In vivo studies further revealed that quercetin promoted fluid secretion in mouse ileum. The modulatory effect of quercetin on CaCC chloirde channels may therefore represent a potential therapeutic strategy for treating CaCC-related diseases like constipation, secretory diarrhea and hypertension. The inverse effects of quercetin on CaCCs provided evidence that ANO1 and intestinal epithelial CaCCs are different calcium-activated chloride channels.
Collapse
Affiliation(s)
- Bo Yu
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University Dalian, China
| | - Yu Jiang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University Dalian, China
| | - Lingling Jin
- College of Basic Medical Sciences, Dalian Medical University Dalian, China
| | - Tonghui Ma
- College of Basic Medical Sciences, Dalian Medical University Dalian, China
| | - Hong Yang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University Dalian, China
| |
Collapse
|
7
|
Sasamoto K, Niisato N, Taruno A, Marunaka Y. Simulation of Cl(-) Secretion in Epithelial Tissues: New Methodology Estimating Activity of Electro-Neutral Cl(-) Transporter. Front Physiol 2015; 6:370. [PMID: 26779025 PMCID: PMC4688368 DOI: 10.3389/fphys.2015.00370] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/20/2015] [Indexed: 12/21/2022] Open
Abstract
Transcellular Cl− secretion is, in general, mediated by two steps; (1) the entry step of Cl− into the cytosolic space from the basolateral space across the basolateral membrane by Cl− transporters, such as Na+-K+-2Cl− cotransporter (NKCC1, an isoform of NKCC), and (2) the releasing step of Cl− from the cytosolic space into the luminal (air) space across the apical membrane via Cl− channels, such as cystic fibrosis transmembrane conductance regulator (CFTR) Cl− channel. Transcellular Cl− secretion has been characterized by using various experimental techniques. For example, measurements of short-circuit currents in the Ussing chamber and patch clamp techniques provide us information on transepithelial ion movements via transcellular pathway, transepithelial conductance, activity (open probability) of single channel, and whole cell currents. Although many investigators have tried to clarify roles of Cl− channels and transporters located at the apical and basolateral membranes in transcellular Cl− secretion, it is still unclear how Cl− channels/transporters contribute to transcellular Cl− secretion and are regulated by various stimuli such as Ca2+ and cAMP. In the present study, we simulate transcellular Cl− secretion using mathematical models combined with electrophysiological measurements, providing information on contribution of Cl− channels/transporters to transcellular Cl− secretion, activity of electro-neutral ion transporters and how Cl− channels/transporters are regulated.
Collapse
Affiliation(s)
- Kouhei Sasamoto
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kyoto, Japan
| | - Naomi Niisato
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of MedicineKyoto, Japan; Department of Health and Sports Sciences, Faculty of Health and Medical Sciences, Kyoto Gakuen UniversityKameoka, Japan; Japan Institute for Food Education and Health, St. Agnes' UniversityKyoto, Japan
| | - Akiyuki Taruno
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kyoto, Japan
| | - Yoshinori Marunaka
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of MedicineKyoto, Japan; Japan Institute for Food Education and Health, St. Agnes' UniversityKyoto, Japan; Department of Bio-Ionomics, Graduate School of Medical Science, Kyoto Prefectural University of MedicineKyoto, Japan
| |
Collapse
|
8
|
Al-Nakkash L, Batia L, Bhakta M, Peterson A, Hale N, Skinner R, Sears S, Jensen J. Stimulation of murine intestinal secretion by daily genistein injections: gender-dependent differences. Cell Physiol Biochem 2011; 28:239-50. [PMID: 21865731 DOI: 10.1159/000331736] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS The effect of daily injections with genistein (naturally occurring phytoestrogen) on intestinal chloride (Cl(-)) secretion was measured with Ussing chamber short circuit current (I(sc), μA/cm(2)), in C57BL/6J male and female mice, using 600 mg/kg genistein/day (600G), 300 mg/kg genistein/day (300G), 150 mg/kg genistein/day (150G) or genistein-free vehicle control (0G) for 1- or 2-weeks. METHODS AND RESULTS Injecting with 600G elicited significant increases in basal I(sc) in females after 1-week (ñ70 μA/cm(2), n=15, p < 0.05) and in males after 2-weeks (ñ80 μA/cm(2), n=5, p < 0.05) compared to their 0G counterparts. Chloride-free ringer significantly reduced basal I(sc) by 65% in 600G males and 72% in 600G females, suggesting that Cl(-) was the major anion comprising the genistein-stimulated secretion. The forskolin-stimulated (10 μM) I(sc) was significantly inhibited by the CFTR chloride channel inhibitors, glibenclamide (500 μM) and CFTR(inh)-172 (100 μM) in 600G males and females, suggesting some contribution by genistein-dependent CFTR-mediated Cl(-) secretion. We found no associated changes in intestinal morphology, nor change in total CFTR protein with 600G. There was a 5% increase in apical/subapical ratio in 600G males compared to controls (no change in females). CONCLUSION These data suggest that male and female mice both exhibit increased Cl- secretion with 600G, however, the mechanisms mediating this are gender-dependent.
Collapse
Affiliation(s)
- Layla Al-Nakkash
- Department of Physiology, Midwestern University, Glendale, AZ 85308, USA.
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Asano J, Niisato N, Nakajima KI, Miyazaki H, Yasuda M, Iwasaki Y, Hama T, Dejima K, Hisa Y, Marunaka Y. Quercetin stimulates Na+/K+/2Cl- cotransport via PTK-dependent mechanisms in human airway epithelium. Am J Respir Cell Mol Biol 2009; 41:688-95. [PMID: 19251944 DOI: 10.1165/rcmb.2008-0338oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We investigated regulatory mechanisms of Cl(-) secretion playing an essential role in the maintenance of surface fluid in human airway epithelial Calu-3 cells. The present study reports that quercetin (a flavonoid) stimulated bumetanide-sensitive Cl(-) secretion with reduction of apical Cl(-) conductance, suggesting that quercetin stimulates Cl(-) secretion by activating an entry step of Cl(-) across the basolateral membrane through Na(+)/K(+)/2Cl(-) cotransporter (NKCC1). To clarify the mechanism stimulating NKCC1 by quercetin, we verified involvement of protein kinase (PK)A, PKC, protein tyrosine kinase (PTK), and cytosolic Ca(2+)-dependent pathways. A PKA inhibitor (PKI-14-22 amide), a PKC inhibitor (Gö 6983) or a Ca(2+) chelating agent did not affect the quercetin-stimulated Cl(-) secretion. On the other hand, a PTK inhibitor (AG18) significantly diminished the stimulatory action of quercetin on Cl(-) secretion without inhibitory effects on apical Cl(-) conductance, suggesting that a PTK-mediated pathway is involved in the stimulatory action of quercetin. The quercetin action on Cl(-) secretion was suppressed with brefeldin A (BFA, an inhibitor of vesicular transport from ER to Golgi), and the BFA-sensitive Cl(-) secretion was not observed in the presence of an epidermal growth factor receptor (EGFR) kinase inhibitor (AG1478), suggesting that quercetin stimulates Cl(-) secretion by causing the EGFR kinase-mediated translocation of NKCC1 or an NKC1-activating factor to the basolateral membrane in human airway epithelial Calu-3 cells. However, the surface density of NKCC1 was not increased by quercetin, but quercetin elevated the activity of NKCC1. These observations indicate that quercetin stimulates Cl(-) secretion by activating NKCC1 via translocation of an NKCC1-activating factor through an EGFR kinase-dependent pathway.
Collapse
Affiliation(s)
- Junji Asano
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wu D, Hu Z. Rutaecarpine induces chloride secretion across rat isolated distal colon. J Pharmacol Exp Ther 2008; 325:256-66. [PMID: 18187619 DOI: 10.1124/jpet.107.131961] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The present study evaluated the effect of rutaecarpine (Rut) on Cl(-) secretion across rat distal colonic mucosa. Basolateral application of Rut elicited an increase in short-circuit current (I(SC)) response in a concentration-dependent manner. Evidence that Rut-stimulated I(SC) was due to Cl(-) secretion is based on 1) inhibition of current by bumetanide; 2) Cl(-) channel blockers diphenylamine-2-carboxylate, 5-nitro-2-(3-phenylpropylamino)-benzoic acid, and glibenclamide; and 3) removal of Cl(-) ions in bath solution. Determination of neurogenic blockers on Rut-induced I(SC) indicated that pretreatment of tissues with tetrodotoxin or indomethacin, but not atropine or hexamethonium, inhibited Rut-induced response. Treatment with Rut led to release and synthesis of prostaglandin E(2) in rat colonic mucosa. Rut-stimulated I(SC) was markedly reduced by pretreatment with MDL-12,330A [cis-N-[2-phenylcyclopentyl]-azacyclotridec-1-en-2-amine] and N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide (H-89), but not with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester, bisindolylmaleimide, and thapsigargin. Elimination of the extracellular Ca(2+) also did not alter Rut response. Rut treatment resulted in the increase in intracellular cAMP levels and the activation of protein kinase A. Depolarizing the basolateral membrane with high K(+) showed that Rut-stimulated apical Cl(-) current was largely prevented by cystic fibrosis transmembrane conductance regulator (CFTR) inhibitors. Permeabilizing apical membrane with nystatin revealed that Rut-stimulated basolateral K(+) current was specifically inhibited by Ba(2+) ions and chromanol 293B. The evidence derived from present study suggests that Rut-stimulated Cl(-) secretion is mediated by generation of endogenous prostaglandin E(2) and that it also involves the stimulation of cAMP and protein kinase A pathways, which subsequently lead to the activation of apical Cl(-) channels, mostly the CFTR and basolateral cAMP-dependent K(+) channels.
Collapse
Affiliation(s)
- DaZheng Wu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Rd., Zhangjiang Hi-tech Park, Shanghai 201203, People's Republic of China.
| | | |
Collapse
|
11
|
Trischitta F, Faggio C. Effect of the flavonol quercetin on ion transport in the isolated intestine of the eel, Anguilla anguilla. Comp Biochem Physiol C Toxicol Pharmacol 2006; 143:17-22. [PMID: 16466967 DOI: 10.1016/j.cbpc.2005.11.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Revised: 11/15/2005] [Accepted: 11/19/2005] [Indexed: 11/19/2022]
Abstract
Flavonoids are phenolic compounds used in fish diet formulations for the control of sex differentiation. Consequently it is of interest to know their effects on fish gastrointestinal mucosa that is the first target of these substances after food intake. We studied the effects of the flavonol quercetin on the transepithelial electrical parameters of the isolated intestine of the eel, Anguilla anguilla, by employing Ussing chamber technique. We showed that luminal quercetin (10(-4) M) reduced the serosa negative I(sc) (short circuit current) due to a net Cl- absorption in the control conditions. The reduction of I(sc) was blocked by luminal glybenclamide (10(-4) M). The I(sc) response to the flavonoid was also observed when HCO3- was omitted from the perfusion solutions. The effect of quercetin was nullified by TMB8, a blocker of Ca2+ release from intracellular stores, and was strongly reduced by trifluoroperazine, an inhibitor of calmodulin actions, while it was unmodified by experimental manoeuvres able to alter cytosolic cAMP concentrations. These results suggest that mucosal quercetin stimulated Cl- but not HCO3- secretion; the secretory activity appears to be dependent on the Ca2+-calmodulin system, independent of the cAMP pathway. In addition, this study showed that flavonol mimics the effect of carbachol on the tissue. This conclusion is supported by the following observations: 1) noradrenalin reversed the reduction of I(sc) produced by both carbachol and quercetin; 2) the flavonol was ineffective in tissues in which the muscarinic agonist had already elicited its maximal reduction of I(sc).
Collapse
Affiliation(s)
- Francesca Trischitta
- Dipartimento di Fisiologia Generale e Farmacologia, Università di Messina, Salita Sperone, 31-98166 Sant'Agata-Messina, Italy.
| | | |
Collapse
|
12
|
Halm ST, Liao T, Halm DR. Distinct K+ conductive pathways are required for Cl- and K+ secretion across distal colonic epithelium. Am J Physiol Cell Physiol 2006; 291:C636-48. [PMID: 16641164 DOI: 10.1152/ajpcell.00557.2005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Secretion of Cl(-) and K(+) in the colonic epithelium operates through a cellular mechanism requiring K(+) channels in the basolateral and apical membranes. Transepithelial current [short-circuit current (I(sc))] and conductance (G(t)) were measured for isolated distal colonic mucosa during secretory activation by epinephrine (Epi) or PGE(2) and synergistically by PGE(2) and carbachol (PGE(2) + CCh). TRAM-34 at 0.5 microM, an inhibitor of K(Ca)3.1 (IK, Kcnn4) K(+) channels (H. Wulff, M. J. Miller, W. Hänsel, S. Grissmer, M. D. Cahalan, and K. G. Chandy. Proc Natl Acad Sci USA 97: 8151-8156, 2000), did not alter secretory I(sc) or G(t) in guinea pig or rat colon. The presence of K(Ca)3.1 in the mucosa was confirmed by immunoblot and immunofluorescence detection. At 100 microM, TRAM-34 inhibited I(sc) and G(t) activated by Epi ( approximately 4%), PGE(2) ( approximately 30%) and PGE(2) + CCh ( approximately 60%). The IC(50) of 4.0 microM implicated involvement of K(+) channels other than K(Ca)3.1. The secretory responses augmented by the K(+) channel opener 1-EBIO were inhibited only at a high concentration of TRAM-34, suggesting further that K(Ca)3.1 was not involved. Sensitivity of the synergistic response (PGE(2) + CCh) to a high concentration TRAM-34 supported a requirement for multiple K(+) conductive pathways in secretion. Clofilium (100 microM), a quaternary ammonium, inhibited Cl(-) secretory I(sc) and G(t) activated by PGE(2) ( approximately 20%) but not K(+) secretion activated by Epi. Thus Cl(-) secretion activated by physiological secretagogues occurred without apparent activity of K(Ca)3.1 channels but was dependent on other types of K(+) channels sensitive to high concentrations of TRAM-34 and/or clofilium.
Collapse
Affiliation(s)
- Susan Troutman Halm
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435, USA
| | | | | |
Collapse
|
13
|
Nakano T, Inoue H, Fukuyama S, Matsumoto K, Matsumura M, Tsuda M, Matsumoto T, Aizawa H, Nakanishi Y. Niflumic acid suppresses interleukin-13-induced asthma phenotypes. Am J Respir Crit Care Med 2006; 173:1216-21. [PMID: 16528019 DOI: 10.1164/rccm.200410-1420oc] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE Chloride channels have been implicated in the regulation of mucus production in epithelial cells. Expression of hCLCA1, a calcium-activated chloride channel, has been reported to be increased in the airway epithelium of patients with asthma. Interleukin (IL)-13 induces the cardinal features of bronchial asthma, and glucocorticoids are not sufficient to suppress IL-13-induced airway hyperresponsiveness or goblet cell hyperplasia. OBJECTIVES We studied the effects of chloride channel inhibitors in IL-13-induced asthma. METHODS The effects of niflumic acid (NA), a relatively specific blocker of calcium-activated chloride channel (CLCA), on goblet cell hyperplasia, eosinophil accumulation, and airway hyperresponsiveness were evaluated after IL-13 instillation into the airways. Because IL-13-dependent features rely on JAK/STAT6 signaling, the effect of NA on phosphorylation of JAK2 and STAT6 after IL-13 stimulation was examined in airway epithelial cells in vitro. The expression of the mCLCA family in mouse lung after IL-13 local administration in vivo was analyzed using reverse transcription-polymerase chain reaction. MEASUREMENTS AND MAIN RESULTS Treatment with NA inhibited not only IL-13-induced goblet cell hyperplasia but also airway hyperresponsiveness and eosinophilic infiltration. NA suppressed the eotaxin levels in bronchoalveolar lavage fluids and overexpression of the MUC5AC gene, a marker of goblet cell hyperplasia, in the lung after IL-13 instillation. NA suppressed JAK2 activation, STAT6 activation, and eotaxin expression in epithelial cells. The expression of mCLCA3 (mouse homolog hCLCA1), but not that of other CLCA family members, was up-regulated by IL-13. CONCLUSIONS These findings suggest that a chloride channel inhibitor can control IL-13-mediated airway features at least by suppressing JAK/STAT6 activation.
Collapse
Affiliation(s)
- Takako Nakano
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Blumenstein I, Keserü B, Wolter F, Stein J. The chemopreventive agent resveratrol stimulates cyclic AMP-dependent chloride secretion in vitro. Clin Cancer Res 2006; 11:5651-6. [PMID: 16061885 DOI: 10.1158/1078-0432.ccr-04-2674] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Resveratrol and its analogs are promising cancer chemoprevention agents, currently under investigation in clinical trials. However, patients administered other plant polyphenols experienced severe diarrhea, likely due to an increase in intracellular cyclic AMP (cAMP). Resveratrol itself raises intracellular cAMP levels in breast cancer cells in vitro. Its future use as a cancer chemopreventive agent could therefore be compromised by its severe side effects. The aim of the study was (a) to define the influence of resveratrol on intestinal Cl(-) secretion and (b) to elucidate possible intracellular transduction pathways involved. Resveratrol caused a dose- and time-dependent increase in DeltaIsc in T(84) cells. The specificity of resveratrol was confirmed by using piceatannol 100 mumol/L, the hydroxylated resveratrol analog, which did not alter DeltaIsc. A significant elevation of [cAMP](i) by resveratrol was assessed in T(84) cells. In mouse jejunum, resveratrol induced a time- and dose-dependent increase in DeltaIsc as well. In bilateral Cl(-)-free medium, as well as after inhibition of protein kinase A, resveratrol-induced DeltaIsc was reduced significantly. Preincubation of T(84) cells with butyrate 2 mmol/L (24 and 48 hours) significantly inhibited resveratrol as well as forskolin-induced Cl(-) secretion. In summary, the main mechanism of action of resveratrol in intestinal epithelia is cAMP-induced chloride secretion which can be suppressed by butyrate. It can therefore be suggested that in cancer chemoprevention, both agents should be combined to reduce an undesired side effect such as diarrhea and to benefit from the known agonistic effect of both agents on differentiation of colon cancer cells.
Collapse
Affiliation(s)
- Irina Blumenstein
- Division of Gastroenterology and Clinical Nutrition, 1 Department of Medicine, ZAFES, J.W. Goethe-Universität, Theodor-Stern-Kai 7, Frankfort on the Main, Germany
| | | | | | | |
Collapse
|
15
|
Hendrich AB. Flavonoid-membrane interactions: possible consequences for biological effects of some polyphenolic compounds. Acta Pharmacol Sin 2006; 27:27-40. [PMID: 16364208 DOI: 10.1111/j.1745-7254.2006.00238.x] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Flavonoids are found ubiquitously in higher plants and constitute an important component of the majority of people daily diets. The biological activities of flavonoids cover a very broad spectrum, from anticancer and antibacterial activities through to inhibition of bone resorption. In the present paper, the interactions between flavonoids and lipid bilayers as well as biological membranes and their components are reviewed, with special emphasis on the structure-activity relationships and mechanisms underlying the biological activity of flavonoids.
Collapse
|
16
|
Chen B, Cai J, Song LS, Wang X, Chen Z. Effects of ginkgo biloba extract on cation currents in rat ventricular myocytes. Life Sci 2005; 76:1111-21. [PMID: 15620575 DOI: 10.1016/j.lfs.2004.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2004] [Accepted: 08/12/2004] [Indexed: 11/19/2022]
Abstract
Ginkgo biloba extract (GBE), a valuable natural product for cerebral and cardiovascular diseases, is mainly composed of two classes of constituents: terpene lactones (e.g., ginkgolide A and B, bilobalide) and flavone glycosides (e.g., quercetin and kaempferol). Its electrophysiological action in heart is yet unclear. In the present study, using whole-cell patch clamp technique, we investigated electrophysiological effects of GBE on cation channel currents in ventricular myocytes isolated from rat hearts. We found that GBE 0.01-0.1% inhibited significantly the sodium current (I(Na)), L-type calcium current (I(Ca)) and transient outward potassium current (IK(to)) in a concentration-dependent manner. Surprisingly, its main ingredients, ginkgolide A (GB A), ginkgolide B (GB B) and bilobalide (GB BA) at 0.1 mM did not exhibit any significant effect on these cation channel currents. These results suggested that GBE is a potent non-selective cation channel modulator in cardiaomyocytes. Other constituents (rather than GB A, GB B and GB BA) might be responsible for the observed inhibitory effects of GBE on cation channels.
Collapse
Affiliation(s)
- Biyi Chen
- Section of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| | | | | | | | | |
Collapse
|
17
|
Cermak R, Wolffram S, Kuhn G. Effect of the flavonol quercetin on membrane conductances in rat colonic crypt cells. Eur J Pharmacol 2004; 503:31-4. [PMID: 15496292 DOI: 10.1016/j.ejphar.2004.09.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2004] [Revised: 09/02/2004] [Accepted: 09/07/2004] [Indexed: 10/26/2022]
Abstract
The plant polyphenol quercetin was shown to induce Cl- secretion in rat colon. This study was performed to investigate the alterations of membrane conductances in isolated epithelial cells induced by quercetin. Whole-cell patch-clamp recordings were performed in isolated crypts from rat distal colon. In cells of the crypt basis, quercetin significantly hyperpolarized the membrane potential at concentrations > or =3 microM and increased the K+ conductance without visibly altering the Cl- conductance. Thus, quercetin induces Cl- secretion merely by activation of K+ channels in the colon epithelium.
Collapse
Affiliation(s)
- Rainer Cermak
- Institute of Animal Nutrition and Physiology, Christian-Albrechts-University of Kiel, Olshausenstr. 40, D-24098 Kiel, Germany.
| | | | | |
Collapse
|
18
|
Yue GGL, Yip TWN, Huang Y, Ko WH. Cellular mechanism for potentiation of Ca2+-mediated Cl- secretion by the flavonoid baicalein in intestinal epithelia. J Biol Chem 2004; 279:39310-6. [PMID: 15234961 DOI: 10.1074/jbc.m406787200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Flavonoids belong to a large group of plant polyphenols that are consumed daily in large amounts. Our previous findings have shown that baicalein, a major flavonoid derived from the medicinal herb Scutellariae radix, induces Cl(-) secretion across rat colonic mucosa. The current study examines the effect of baicalein on Cl(-) secretion in human colonic epithelial (T84) cells and its interaction with Ca(2+)- and cAMP-dependent secretagogues. We have employed a technique that allows concurrent monitoring of short-circuit current (I(SC)) and [Ca(2+)](i) in polarized epithelium. Basolateral application of baicalein induced a concentration-dependent increase in I(SC). The increase in I(SC) was because of Cl(-) secretion and was not accompanied by any discernible increase in [Ca(2+)](i). Baicalein acted synergistically with Ca(2+)- but not cAMP-dependent secretagogues. In the presence of baicalein, the carbachol and histamine induced increases in I(SC) that were markedly potentiated while increases in [Ca(2+)](i) were not significantly enhanced. Baicalein treatment uncoupled Cl(-) secretion from inhibitory effects normally generated by muscarinic activation. Baicalein treatment also resulted in increased cAMP content and activated PKA activity. Nystatin permeabilization studies revealed that baicalein stimulated an apical Cl(-) current but did not activate any basolateral K(+) current. These data suggest that baicalein potentiates Ca(2+)-mediated Cl(-) secretion through a signaling pathway involving cAMP and protein kinase A, most likely through the cystic fibrosis transmembrane conductance regulator in the apical membrane.
Collapse
Affiliation(s)
- Grace Gar-Lee Yue
- Department of Physiology, The Chinese University of Hong Kong, Shatin, N.T. Hong Kong, China
| | | | | | | |
Collapse
|