Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 321(6): C932-C946, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34644122

ABSTRACT

Chloride transport across cell membranes is broadly involved in epithelial fluid transport, cell volume and pH regulation, muscle contraction, membrane excitability, and organellar acidification. The human genome encodes at least 53 chloride-transporting proteins with expression in cell plasma or intracellular membranes, which include chloride channels, exchangers, and cotransporters, some having broad anion specificity. Loss-of-function mutations in chloride transporters cause a wide variety of human diseases, including cystic fibrosis, secretory diarrhea, kidney stones, salt-wasting nephropathy, myotonia, osteopetrosis, hearing loss, and goiter. Although impactful advances have been made in the past decade in drug treatment of cystic fibrosis using small molecule modulators of the defective cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel, other chloride channels and solute carrier proteins (SLCs) represent relatively underexplored target classes for drug discovery. New opportunities have emerged for the development of chloride transport modulators as potential therapeutics for secretory diarrheas, constipation, dry eye disorders, kidney stones, polycystic kidney disease, hypertension, and osteoporosis. Approaches to chloride transport-targeted drug discovery are reviewed herein, with focus on chloride channel and exchanger classes in which recent preclinical advances have been made in the identification of small molecule modulators and in proof of concept testing in experimental animal models.


Subject(s)
Antiporters/drug effects , Chloride Channels/drug effects , Chlorides/metabolism , Drug Design , Drug Discovery , Membrane Transport Modulators/pharmacology , Animals , Antiporters/genetics , Antiporters/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Ion Transport , Kinetics , Membrane Transport Modulators/chemistry , Mutation , Sulfate Transporters/drug effects , Sulfate Transporters/genetics , Sulfate Transporters/metabolism
2.
Nat Commun ; 12(1): 172, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420032

ABSTRACT

The dimeric transporter, EmrE, effluxes polyaromatic cationic drugs in a proton-coupled manner to confer multidrug resistance in bacteria. Although the protein is known to adopt an antiparallel asymmetric topology, its high-resolution drug-bound structure is so far unknown, limiting our understanding of the molecular basis of promiscuous transport. Here we report an experimental structure of drug-bound EmrE in phospholipid bilayers, determined using 19F and 1H solid-state NMR and a fluorinated substrate, tetra(4-fluorophenyl) phosphonium (F4-TPP+). The drug-binding site, constrained by 214 protein-substrate distances, is dominated by aromatic residues such as W63 and Y60, but is sufficiently spacious for the tetrahedral drug to reorient at physiological temperature. F4-TPP+ lies closer to the proton-binding residue E14 in subunit A than in subunit B, explaining the asymmetric protonation of the protein. The structure gives insight into the molecular mechanism of multidrug recognition by EmrE and establishes the basis for future design of substrate inhibitors to combat antibiotic resistance.


Subject(s)
Antiporters/chemistry , Antiporters/drug effects , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/drug effects , Lipid Bilayers/chemistry , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Biological Transport/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/metabolism , Molecular Dynamics Simulation , Protein Conformation
3.
Curr Opin Support Palliat Care ; 14(2): 107-111, 2020 06.
Article in English | MEDLINE | ID: mdl-32349095

ABSTRACT

PURPOSE OF REVIEW: The treatment of cancer-induced bone pain (CIBP) has been proven ineffective and relies heavily on opioids, the target of highly visible criticism for their negative side effects. Alternative therapeutic agents are needed and the last few years have brought promising results, detailed in this review. RECENT FINDINGS: Cysteine/glutamate antiporter system, xc, cannabinoids, kappa opioids, and a ceramide axis have all been shown to have potential as novel therapeutic targets without the negative effects of opioids. SUMMARY: Review of the most recent and promising studies involving CIBP, specifically within murine models. Cancer pain has been reported by 30-50% of all cancer patients and even more in late stages, however the standard of care is not effective to treat CIBP. The complicated and chronic nature of this type of pain response renders over the counter analgesics and opioids largely ineffective as well as difficult to use due to unwanted side effects. Preclinical studies have been standardized and replicated while novel treatments have been explored utilizing various alternative receptor pathways: cysteine/glutamate antiporter system, xc, cannabinoid type 1 receptor, kappa opioids, and a ceramide axis sphingosine-1-phosphate/sphingosine-1-phosphate receptor 1.


Subject(s)
Bone and Bones/physiopathology , Cancer Pain/drug therapy , Animals , Antiporters/drug effects , Antiporters/metabolism , Bone and Bones/innervation , Cannabinoids/therapeutic use , Disease Models, Animal , Humans , Mice , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, kappa/metabolism , Sphingosine-1-Phosphate Receptors/drug effects , Sphingosine-1-Phosphate Receptors/metabolism
4.
Invest Ophthalmol Vis Sci ; 59(10): 4258-4267, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30140924

ABSTRACT

Purpose: SLC4A11 is a plasma membrane protein of corneal endothelial cells. Some mutations of the SLC4A11 gene result in SLC4A11 protein misfolding and failure to mature to the plasma membrane. This gives rise to some cases of Fuchs' endothelial corneal dystrophy (FECD) and congenital hereditary endothelial dystrophy (CHED). We screened ophthalmic nonsteroidal anti-inflammatory drugs (NSAIDs) for their ability to correct SLC4A11 folding defects. Methods: Five ophthalmic NSAIDs were tested for their therapeutic potential in some genetic corneal dystrophy patients. HEK293 cells expressing CHED and FECD-causing SLC4A11 mutants were grown on 96-well dishes in the absence or presence of NSAIDs. Ability of NSAIDs to correct mutant SLC4A11 cell-surface trafficking was assessed with a bioluminescence resonance energy transfer (BRET) assay and by confocal microscopy. The ability of mutant SLC4A11-expressing cells to mediate water flux (SLC4A11 mediates water flux across the corneal endothelial cell basolateral membrane as part of the endothelial water pump) was measured upon treatment with ophthalmic NSAIDs. Results: BRET-assays revealed significant rescue of SLC4A11 mutants to the cell surface by 4 of 5 NSAIDs tested. The NSAIDs, diclofenac and nepafenac, were effective in moving endoplasmic reticulum-retained missense mutant SLC4A11 to the cell surface, as measured by confocal immunofluorescence. Among intracellular-retained SLC4A11 mutants, 20 of 30 had significant restoration of cell surface abundance upon treatment with diclofenac. Diclofenac restored mutant SLC4A11 water flux activity to the level of wild-type SLC4A11 in some cases. Conclusions: These results encourage testing diclofenac eye drops as a treatment for corneal dystrophy in patients whose disease is caused by some SLC4A11 missense mutations.


Subject(s)
Anion Transport Proteins/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiporters/drug effects , Biological Transport/drug effects , Corneal Dystrophies, Hereditary/drug therapy , Corneal Dystrophies, Hereditary/genetics , Diclofenac/pharmacology , Endothelium, Corneal/drug effects , Water/metabolism , Anion Transport Proteins/chemistry , Anion Transport Proteins/genetics , Antiporters/chemistry , Antiporters/genetics , Cell Membrane/drug effects , Cells, Cultured , Endothelial Cells/drug effects , HEK293 Cells , Humans , Mutation, Missense , Protein Folding
5.
J Biochem Mol Toxicol ; 31(4)2017 Apr.
Article in English | MEDLINE | ID: mdl-27891704

ABSTRACT

We hypothesized that genistein could affect the chloride (Cl- ) and bicarbonate (HCO3- ) secretory mechanisms in uterus. Ovariectomized female rats were given estradiol or estradiol plus progesterone with 25, 50, or 100 mg/kg/day genistein. Following completion of the treatment, uterine fluid Cl- and HCO3- concentrations were determined by in vivo uterine perfusion. Uteri were subjected for molecular biological analysis (Western blot, qPCR, and immunohistochemistry) to detect levels of expression of Cystic Fibrosis transmembrane regulator (CFTR), Cl- /HCO3- exchanger (SLC26a6), Na+ /HCO3- cotransporter (SLC4a4), and estrogen receptor (ER)-α and ß. Coadministration of genistein resulted in decrease in Cl- and HCO3- concentrations and expression of CFTR, SLC26a6, SLC4a4, and ER-α and ER-ß in the uteri of estradiol-treated rats. In estradiol plus progesterone-treated rats, a significant increase in the above parameters were observed following high-dose genistein treatment except for the SLC24a4 level. In conclusion, genistein-induced changes in the uterus could affect the reproductive processes that might result in infertility.


Subject(s)
Bicarbonates/metabolism , Chlorides/metabolism , Estrogens/pharmacology , Genistein/pharmacology , Uterus/drug effects , Animals , Antiporters/drug effects , Antiporters/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Female , Gene Expression Regulation , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Sodium-Bicarbonate Symporters/drug effects , Sodium-Bicarbonate Symporters/genetics , Sulfate Transporters , Uterus/metabolism
6.
J Cyst Fibros ; 14(5): 561-70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26115565

ABSTRACT

Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause a characteristic defect in epithelial ion transport that plays a central role in the pathogenesis of cystic fibrosis (CF). Hence, pharmacological correction of this ion transport defect by targeting of mutant CFTR, or alternative ion channels that may compensate for CFTR dysfunction, has long been considered as an attractive approach to a causal therapy of this life-limiting disease. The recent introduction of the CFTR potentiator ivacaftor into the therapy of a subgroup of patients with specific CFTR mutations was a major milestone and enormous stimulus for seeking effective ion transport modulators for all patients with CF. In this review, we discuss recent breakthroughs and setbacks with CFTR modulators designed to rescue mutant CFTR including the common mutation F508del. Further, we examine the alternative chloride channels TMEM16A and SLC26A9, as well as the epithelial sodium channel ENaC as alternative targets in CF lung disease, which remains the major cause of morbidity and mortality in patients with CF. Finally, we will focus on the hurdles that still need to be overcome to make effective ion transport modulation therapies available for all patients with CF irrespective of their CFTR genotype.


Subject(s)
Aminophenols/therapeutic use , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis , Quinolones/therapeutic use , Anoctamin-1 , Antiporters/drug effects , Antiporters/metabolism , Chloride Channels/drug effects , Chloride Channels/metabolism , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/metabolism , Humans , Ion Transport/genetics , Mutation/drug effects , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Sulfate Transporters
7.
Physiol Res ; 64(1): 71-8, 2015.
Article in English | MEDLINE | ID: mdl-25194133

ABSTRACT

Previous studies in our laboratory reported L-malate as a free radical scavenger in aged rats. To investigate the antioxidant mechanism of L-malate in the mitochondria, we analyzed the change in gene expression of two malate-aspartate shuttle (MAS)-related carried proteins (AGC, aspartate/glutamate carrier and OMC, oxoglutarate/malate carrier) in the inner mitochondrial membrane, and three antioxidant enzymes (CAT, SOD, and GSH-Px) in the mitochondria. The changes in gene expression of these proteins and enzymes were examined by real-time RT-PCR in the heart and liver of aged rats treated with L-malate. L-malate was orally administered in rats continuously for 30 days using a feeding atraumatic needle. We found that the gene expression of OMC and GSH-Px mRNA in the liver increased by 39 % and 38 %, respectively, in the 0.630 g/kg L-malate treatment group than that in the control group. The expression levels of SOD mRNA in the liver increased by 39 %, 56 %, and 78 % in the 0.105, 0.210, and 0.630 g/kg L-malate treatment groups, respectively. No difference were observed in the expression levels of AGC, OMC, CAT, SOD, and GSH-Px mRNAs in the heart of rats between the L-malate treatment and control groups. These results predicted that L-malate may increase the antioxidant capacity of mitochondria by enhancing the expression of mRNAs involved in the MAS and the antioxidant enzymes.


Subject(s)
Aging/metabolism , Antioxidants/metabolism , Antioxidants/pharmacology , Enzymes/metabolism , Liver/drug effects , Malates/pharmacology , Membrane Transport Proteins/drug effects , Age Factors , Aging/genetics , Amino Acid Transport Systems, Acidic/drug effects , Amino Acid Transport Systems, Acidic/metabolism , Animals , Antiporters/drug effects , Antiporters/metabolism , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Catalase/metabolism , Enzymes/genetics , Gene Expression Regulation, Enzymologic , Glutathione Peroxidase/metabolism , Liver/enzymology , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/enzymology , Mitochondrial Proteins , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism , Up-Regulation
8.
Gut ; 63(10): 1587-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24440986

ABSTRACT

BACKGROUND: ClC-3 channel/antiporter plays a critical role in a variety of cellular activities. ClC-3 has been detected in the ileum and colon. OBJECTIVE: To determine the functions of ClC-3 in the gastrointestinal tract. DESIGN: After administration of dextran sulfate sodium (DSS) or 2,4,6-trinitrobenzenesulfonic acid (TNBS), intestines from ClC-3-/- and wild-type mice were examined by histological, cellular, molecular and biochemical approaches. ClC-3 expression was determined by western blot and immunostaining. RESULTS: ClC-3 expression was reduced in intestinal tissues from patients with UC or Crohn's disease and from mice treated with DSS. Genetic deletion of ClC-3 increased the susceptibility of mice to DSS- or TNBS-induced experimental colitis and prevented intestinal recovery. ClC-3 deficiency promoted DSS-induced apoptosis of intestinal epithelial cells through the mitochondria pathway. ClC-3 interacts with voltage-dependent anion channel 1, a key player in regulation of mitochondria cytochrome c release, but DSS treatment decreased this interaction. In addition, lack of ClC-3 reduced the numbers of Paneth cells and impaired the expression of antimicrobial peptides. These alterations led to dysfunction of the epithelial barrier and invasion of commensal bacteria into the mucosa. CONCLUSIONS: A defect in ClC-3 may contribute to the pathogenesis of IBD by promoting intestinal epithelial cell apoptosis and Paneth cell loss, suggesting that modulation of ClC-3 expression might be a new strategy for the treatment of IBD.


Subject(s)
Antiporters/metabolism , Chloride Channels/physiology , Colitis, Ulcerative/metabolism , Crohn Disease/metabolism , Gastrointestinal Tract/metabolism , Paneth Cells/pathology , Animals , Antiporters/drug effects , Apoptosis , Blotting, Western , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Crohn Disease/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/pathology , Humans , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Trinitrobenzenesulfonic Acid/toxicity
9.
AAPS J ; 15(2): 299-307, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23212563

ABSTRACT

Nicotine, the main tobacco alkaloid leading to smoking dependence, rapidly crosses the blood-brain barrier (BBB) to become concentrated in the brain. Recently, it has been shown that nicotine interacts with some organic cation transporters (OCT), but their influence at the BBB has not yet been assessed in vivo. In this study, we characterized the transport of nicotine at the mouse luminal BBB by in situ brain perfusion. Its influx was saturable and followed the Michaelis-Menten kinetics (K(m)=2.60 mM, V(max)=37.60 nmol/s/g at pH 7.40). At its usual micromolar concentrations in the plasma, most (79%) of the net transport of nicotine at the BBB was carrier-mediated, while passive diffusion accounted for 21%. Studies on knockout mice showed that the OCT Oct1-3, P-gp, and Bcrp did not alter [(3)H]-nicotine transport at the BBB. Neither did inhibiting the transporters Mate1, Octn, or Pmat. The in vivo manipulation of intracellular and/or extracellular pH, the chemical inhibition profile, and the trans-stimulation experiments demonstrated that the nicotine transporter at the BBB shared the properties of the clonidine/proton antiporter. The molecular features of this proton-coupled antiporter have not yet been identified, but it also transports diphenhydramine and tramadol and helps nicotine cross the BBB at a faster rate and to a greater extent. The pharmacological inhibition of this nicotine/proton antiporter could represent a new strategy to reduce nicotine uptake by the brain and thus help curb addiction to smoking.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antiporters/metabolism , Blood-Brain Barrier/metabolism , Nicotine/metabolism , Organic Cation Transport Proteins/metabolism , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/genetics , Animals , Antiporters/drug effects , Binding, Competitive , Biological Transport , Blood-Brain Barrier/drug effects , Diffusion , Diphenhydramine/metabolism , Hydrogen-Ion Concentration , Kinetics , Male , Mice , Mice, Knockout , Models, Biological , Nonlinear Dynamics , Organic Cation Transport Proteins/deficiency , Organic Cation Transport Proteins/drug effects , Organic Cation Transport Proteins/genetics , Perfusion , Tramadol/metabolism
10.
Am J Respir Cell Mol Biol ; 45(4): 684-91, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21474433

ABSTRACT

To investigate the effects of capsaicinoids on airway anion transporters, we recorded and analyzed transepithelial currents in human airway epithelial Calu-3 cells. Application of capsaicin (100 µM) attenuated vectorial anion transport, estimated as short-circuit currents (I(SC)), before and after stimulation by forskolin (10 µM) with concomitant reduction of cytosolic cyclic AMP (cAMP) levels. The capsaicin-induced inhibition of I(SC) was also observed in the response to 8-bromo-cAMP (1 mM, a cell-permeable cAMP analog) and 3-isobutyl-1-methylxanthine (1 mM, an inhibitor of phosphodiesterases). The capsaicin-induced inhibition of I(SC) was attributed to suppression of bumetanide (an inhibitor of the basolateral Na(+)-K(+)-2 Cl(-) cotransporter 1)- and 4,4'-dinitrostilbene-2,2'-disulfonic acid (an inhibitor of basolateral HCO(3)(-)-dependent anion transporters)-sensitive components, which reflect anion uptake via basolateral cAMP-dependent anion transporters. In contrast, capsaicin potentiated apical Cl(-) conductance, which reflects conductivity through the cystic fibrosis transmembrane conductance regulator, a cAMP-regulated Cl(-) channel. All these paradoxical effects of capsaicin were mimicked by capsazepine. Forskolin application also increased phosphorylated myosin phosphatase target subunit 1, and the phosphorylation was prevented by capsaicin and capsazepine, suggesting that these capsaicinoids assume aspects of Rho kinase inhibitors. We also found that the increments in apical Cl(-) conductance were caused by conventional Rho kinase inhibitors, Y-27632 (20 µM) and HA-1077 (20 µM), with selective inhibition of basolateral Na(+)-K(+)-2 Cl(-) cotransporter 1. Collectively, capsaicinoids inhibit cAMP-mediated anion transport through down-regulation of basolateral anion uptake, paradoxically accompanied by up-regulation of apical cystic fibrosis transmembrane conductance regulator-mediated anion conductance. The latter is mediated by inhibition of Rho-kinase, which is believed to interact with actin cytoskeleton.


Subject(s)
Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cyclic AMP/metabolism , Epithelial Cells/drug effects , Membrane Transport Proteins/drug effects , Protein Kinase Inhibitors/pharmacology , Respiratory Mucosa/drug effects , rho-Associated Kinases/antagonists & inhibitors , Adenylyl Cyclases/metabolism , Anion Transport Proteins/drug effects , Anion Transport Proteins/metabolism , Antiporters/drug effects , Antiporters/metabolism , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Activators/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/enzymology , Humans , Ion Transport , Membrane Potentials , Membrane Transport Proteins/metabolism , Patch-Clamp Techniques , Respiratory Mucosa/enzymology , SLC4A Proteins , Sodium-Bicarbonate Symporters/drug effects , Sodium-Bicarbonate Symporters/metabolism , Sodium-Potassium-Chloride Symporters/drug effects , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 2 , rho-Associated Kinases/metabolism
11.
Trends Biochem Sci ; 32(6): 252-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17452106

ABSTRACT

When biochemistry meets structural biology a more complete understanding of the mechanism of biological macromolecules is usually achieved. Several high-resolution structures of ion-coupled transporters have enriched the understanding of mechanisms of substrate recognition, translocation and coupling of substrate fluxes. However, two X-ray structures of EmrE, the smallest ion-coupled multi-drug transporter, raised questions over the veracity of the structural model and represented a cautionary tale about the difficulty of determining the 3D structures of membrane proteins and the dangers of ignoring biochemical results. The 3D structures of EmrE have since been retracted because of faulty software, but the suggestion that the protomers in the dimer are in an antiparallel topological orientation sparked controversy that is still ongoing.


Subject(s)
Antiporters/chemistry , Escherichia coli Proteins/chemistry , Antiporters/drug effects , Crystallography, X-Ray , Detergents/pharmacology , Dimerization , Escherichia coli Proteins/drug effects , Membrane Proteins/chemistry , Models, Molecular , Protein Structure, Secondary/drug effects , Retraction of Publication as Topic , Software
12.
Phytother Res ; 19(7): 624-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16161025

ABSTRACT

In intact microsomes, quercetin 3-O-alpha-(2''-galloyl)rhamnoside (QGR) inhibits glucose-6-phosphatase (G-6-Pase) in a concentration-dependent manner. QGR increased the G-6-Pase K(m) for glucose-6-phosphate without change in the V(max). The flavonol did not change the kinetic parameters of disrupted microsomal G-6-Pase or intact or disrupted microsomal G-6-Pase pyrophosphatase (PPase) activity. This result allowed the conclusion that QGR competitively inhibits the glucose-6-phosphate (G-6-P) transporter (T1) without affecting the catalytic subunit or the phosphate/pyrophosphate transporter (T2) of the G-6-Pase system.QGR strongly inhibits the neoglucogenic capacity of rat liver slices incubated in a Krebs-Ringer bicarbonate buffer, supplemented with lactate and oleate saturated albumin. The QGR G-6-Pase inhibition might explain the decrease in the liver slice neoglucogenic capacity and, in turn, could reduce glucose levels in diabetic patients.


Subject(s)
Bauhinia , Enzyme Inhibitors/pharmacology , Gluconeogenesis/drug effects , Glucose-6-Phosphatase/drug effects , Phytotherapy , Plant Extracts/pharmacology , Animals , Antiporters/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Glucose-6-Phosphatase/antagonists & inhibitors , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Monosaccharide Transport Proteins/drug effects , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Plant Leaves , Quercetin/administration & dosage , Quercetin/pharmacology , Quercetin/therapeutic use , Rats , Rats, Sprague-Dawley
13.
Am J Physiol Gastrointest Liver Physiol ; 289(4): G768-78, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16002564

ABSTRACT

Normal human colonic luminal (NH(4)(+)) concentration ([NH(4)(+)]) ranges from approximately 10 to 100 mM. However, the nature of the effects of NH(4)(+) on transport, as well as NH(4)(+) transport itself, in colonic epithelium is poorly understood. We elucidate here the effects of apical NH(4)(+) on cAMP-stimulated Cl(-) secretion in colonic T84 cells. In HEPES-buffered solutions, 10 mM apical NH(4)(+) had no significant effect on cAMP-stimulated current. In contrast, 10 mM apical NH(4)(+) reduced current within 5 min to 61 +/- 4% in the presence of 25 mM HCO(3)(-). Current inhibition was not simply due to an increase in extracellular K(+)-like cations, in that the current magnitude was 95 +/- 5% with 10 mM apical K(+) and 46 +/- 3% with 10 mM apical NH(4)(+) relative to that with 5 mM apical K(+). We previously demonstrated that inhibition of Cl(-) secretion by basolateral NH(4)(+) occurs in HCO(3)(-)-free conditions and exhibits anomalous mole fraction behavior. In contrast, apical NH(4)(+) inhibition of current in HCO(3)(-) buffer did not show anomalous mole fraction behavior and followed the absolute [NH(4)(+)] in K(+)-NH(4)(+) mixtures, where K(+) concentration + [NH(4)(+)] = 10 mM. The apical NH(4)(+) inhibitory effect was not prevented by 100 microM methazolamide, suggesting no role for apical carbonic anhydrase. However, apical NH(4)(+) inhibition of current was prevented by 10 min of pretreatment of the apical surface with 500 microM DIDS, 100 microM 4,4'-dinitrostilbene-2,2'-disulfonic acid (DNDS), or 25 microM niflumic acid, suggesting a role for NH(4)(+) action through an apical anion exchanger. mRNA and protein for the apical anion exchangers SLC26A3 [downregulated in adenoma (DRA)] and SLC26A6 [putative anion transporter (PAT1)] were detected in T84 cells by RT-PCR and Northern and Western blots. DRA and PAT1 appear to associate with CFTR in the apical membrane. We conclude that the HCO(3)(-) dependence of apical NH(4)(+) inhibition of secretion is due to the action of NH(4)(+) on an apical anion exchanger.


Subject(s)
Bicarbonates/metabolism , Colon/metabolism , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/pharmacology , Quaternary Ammonium Compounds/pharmacology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Antiporters/drug effects , Antiporters/metabolism , Blotting, Northern , Blotting, Western , Carbachol/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , Cell Line , Chloride Channels/antagonists & inhibitors , Colon/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Membrane Transport Proteins/metabolism , Muscarinic Agonists/pharmacology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Stilbenes/pharmacology , Sulfate Transporters
14.
J Cardiovasc Electrophysiol ; 16(4): 433-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15828890

ABSTRACT

INTRODUCTION: Deaths secondary to low-energy impacts to the precordium in young individuals (commotio cordis) have been reported with increasing frequency. In a swine model, baseball impacts induce ventricular fibrillation when directed at the center of the left ventricle during the vulnerable portion of repolarization just prior to the T-wave peak. It has been hypothesized that activation of stretch-sensitive channels could be crucial for this electrophysiological phenomenon. In this study, a nonselective stretch-activated cation channel was pharmacologically blocked prior to chest blows to determine whether this channel represents a possible pathway by which commotio cordis events occur. METHODS: In a randomized and blinded experiment, 12 swine (mean 17.1 +/- 2.5 kg) received either 2-g streptomycin intramuscularly (mean serum concentration 115 +/- 18 muM) or sterile water prior to chest impact. Each animal received six precordial impacts with a baseball propelled at 40 mph. RESULTS: There was no significant difference in the frequency of induced VF in the animals administered streptomycin (10 of 19 impacts: 53%) compared to those control animals receiving only sterile water (10 of 31: 32%) (P = 0.15). However, the magnitude of ST segment elevation was less in the streptomycin-treated animals (19 +/- 19 mV) versus controls (61 +/- 46 mV) (P = 0.015). CONCLUSION: Streptomycin did not alter the frequency of ventricular fibrillation in our commotio cordis model, indicating that the stretch-activated channel is not implicated in the genesis of chest blow-induced cardiac arrest. However, streptomycin did reduce ST elevation following impact suggesting that the stretch-activated channel may play a role in ST segment elevation following chest wall blows.


Subject(s)
Antiporters/drug effects , Death, Sudden, Cardiac/etiology , Heart Injuries/complications , Protein Synthesis Inhibitors/pharmacology , Streptomycin/pharmacology , Thoracic Injuries/complications , Animals , Antiporters/metabolism , Disease Models, Animal , Electrocardiography , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Male , Random Allocation , Swine , Ventricular Fibrillation/complications , Ventricular Fibrillation/metabolism , Ventricular Fibrillation/physiopathology , Ventricular Pressure/physiology , Wounds, Nonpenetrating/complications
15.
J Cell Physiol ; 199(3): 434-40, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15095290

ABSTRACT

Regulation of cellular Mg(2+) levels by insulin has been shown in various tissues. However, the mechanisms for hormonal regulation of cellular Mg(2+) have not been well described. We studied the effect of insulin on Na(+)/Mg(2+) exchange in normal human cells, measuring Na(+)/Mg(2+) exchange activity as net total Mg(2+) efflux driven by an inward Na(+) gradient in Mg(2+)-loaded red blood cells (RBCs). Na(+)/Mg(2+) exchange was increased significantly by the addition of 2.4 nmol/L of insulin to the flux medium (from 0.60 +/- 0.06 mmol/L cell x h to 0.75 +/- 0.08 mmol/L cell x h [P = 0.0098, n = 44]). A dose-response curve for the effects of insulin on the exchanger activity gave an estimated EC(50) for insulin of 0.95 +/- 0.31 nmol/L and a V(max) of 0.86 +/- 0.12 mmol/L cell x h (n = 7). Kinetics of the Na(+)/Mg(2+) exchange were characterized by measuring its activity as a function of Mg(2+) and Na(+) concentrations. The K(0.5) for cellular Mg(2+) was not affected by incubation with insulin. However, the K(0.5) for extracellular Na(+) decreased from 69.9 +/- 6.3 to 40.3 +/- 8.4 mol/L (n = 5, P = 0.03) in the presence of insulin. We also studied the effect of wortmannin (WT), a PI 3-kinase inhibitor, on activity of the exchanger. WT significantly blocked the insulin-stimulated Na(+)/Mg(2+) activity (n = 6, P = 0.048), with an IC(50) of 0.5 nmol/L. LY294002, another PI 3-kinase inhibitor, likewise blocked the insulin-stimulated activity of the exchanger. Therefore, insulin regulates cellular Mg(2+) metabolism in part via an increase in the affinity for Na(+) of the Na(+)/Mg(2+) exchange and PI 3-kinase activation, suggesting another role for the PI 3-kinase pathway in insulin-mediated cellular events.


Subject(s)
Antiporters/physiology , Insulin/metabolism , Magnesium/metabolism , Receptor, Insulin/physiology , Sodium/metabolism , Antiporters/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Erythrocytes/drug effects , Erythrocytes/metabolism , Humans , Insulin/pharmacology , Phosphoinositide-3 Kinase Inhibitors
16.
Plant J ; 37(4): 578-88, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14756768

ABSTRACT

During drought, the plant hormone abscisic acid (ABA) induces rapid stomatal closure and in turn reduces transpiration. Stomatal closure is accompanied by large ion fluxes across the plasma membrane, carried by K+ and anion channels. We recorded changes in the activity of these channels induced by ABA, for guard cells of intact Vicia faba plants. Guard cells in their natural environment were impaled with double-barrelled electrodes, and ABA was applied via the leaf surface. In 45 out of 85 cells tested, ABA triggered a transient depolarization of the plasma membrane. In these cells, the membrane potential partially recovered in the presence of ABA; however, a full recovery of the membrane potentials was only observed after removal of ABA. Repetitive ABA responses could be evoked in single cells, but the magnitude of the response varied from one hormone application to the other. The transient depolarization correlated with the activation of anion channels, which peaked 5 min after introduction of the stimulus. In guard cells with a moderate increase in plasma membrane conductance (DeltaG < 5 nS), ABA predominantly activated voltage-independent (slow (S)-type) anion channels. During strong responses (DeltaG > 5 nS), however, ABA activated voltage-dependent (rapid (R)-type) in addition to S-type anion channels. We conclude that the combined activation of these two channel types leads to the transient depolarization of guard cells. The nature of this ABA response correlates with the transient extrusion of Cl- from guard cells and a rapid but confined reduction in stomatal aperture.


Subject(s)
Abscisic Acid/pharmacology , Plant Epidermis/drug effects , Plant Growth Regulators/pharmacology , Potassium Channels/drug effects , Vicia faba/drug effects , Antiporters/drug effects , Antiporters/metabolism , Cell Membrane/drug effects , Cell Membrane/physiology , Membrane Potentials/drug effects , Models, Biological , Plant Epidermis/cytology , Plant Epidermis/metabolism , Potassium Channels/metabolism , Vicia faba/cytology , Vicia faba/metabolism
17.
Biochemistry (Mosc) ; 68(11): 1247-51, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14640968

ABSTRACT

The role of Cl- transport across the plasma membrane was studied in an early step of pollen grain germination in tobacco Nicotiana tabacum L. The Cl- channel blockers, 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB) and niflumic acid, completely suppress the germination with IC(50) approximately 8 micro M. At this concentration NPPB reduces the rate of Cl- efflux out of pollen grain by 1.8-fold in the interval 5-12 min, and niflumic acid reduces the rate 1.2-fold. 4,4;-Diisothiocyanatostilbene-2,2;-disulfonic acid, a known inhibitor of Cl- channels and antiporters, completely suppresses germination as well (IC(50) = 240 micro M), but has no effect on the rate of Cl- efflux. Inhibitors of chloride co-transporters, such as furosemide, bumetanide, and bis(1,3-dibutylbarbituric acid)pentamethine oxonol, suppress the germination by less than 50%. This set of data suggests that NPPB-sensitive anion channels are involved in the activation of pollen grains in the early stage of germination.


Subject(s)
Chloride Channels/antagonists & inhibitors , Germination/drug effects , Nicotiana/physiology , Pollen/physiology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Antiporters/drug effects , Barbiturates/pharmacology , Bumetanide/pharmacology , Furosemide/pharmacology , Ion Transport/drug effects , Isoxazoles/pharmacology , Niflumic Acid/pharmacology , Nitrobenzoates/pharmacology
18.
Leukemia ; 17(4): 746-50, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12682632

ABSTRACT

We have previously reported that prodigiosin (2-methyl-3-pentyl-6-methoxyprodigiosene) induces apoptosis in human hematopoietic cancer cell lines with no marked toxicity in nonmalignant cell lines. In this study, we demonstrate that prodigiosin induces apoptosis of B-cell chronic lymphocytic leukemia (B-CLL) cells (n=32 patients). The dose-response for the cytotoxic effect of prodigiosin was analyzed in cells from 12 patients showing an IC(50) of 116+/-25 nM. Prodigiosin induced apoptosis of B-CLL cells through caspase activation. We also analyzed the cytotoxic effect of prodigiosin in T cells from B-CLL samples and no differences were observed with respect to leukemia cells. This is the first report showing that prodigiosin induces apoptosis in human primary cancer cells.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplastic Stem Cells/drug effects , Prodigiosin/pharmacology , T-Lymphocytes/drug effects , Antiporters/drug effects , B-Lymphocytes/chemistry , B-Lymphocytes/pathology , Caspases/metabolism , Enzyme Activation/drug effects , Humans , Ion Transport/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/chemistry , Neoplastic Stem Cells/pathology , T-Lymphocytes/chemistry , T-Lymphocytes/pathology , Tumor Cells, Cultured/drug effects , Tumor Suppressor Protein p53/analysis
19.
Antimicrob Agents Chemother ; 47(1): 27-33, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12499164

ABSTRACT

Of the six putative small multidrug resistance (SMR) family proteins of Pseudomonas aeruginosa, a protein encoded by the PA4990 gene (emrE(Pae)) shows the highest identity to the well-characterized EmrE efflux transporter of Escherichia coli. Reverse transcription-PCR confirmed the expression of emrE(Pae) in the wild-type strain of P. aeruginosa. Using isogenic emrE(Pae), mexAB-oprM, and/or mexB deletion mutants, the contributions of the EmrE protein and the MexAB-OprM efflux system to drug resistance in P. aeruginosa were assessed by a drug susceptibility test carried out in a low-ionic-strength medium, Difco nutrient broth. We found that EmrE(Pae) contributed to intrinsic resistance not only to ethidium bromide and acriflavine but also to aminoglycosides. In this low-ionic-strength medium, MexAB-OprM was also shown to contribute to aminoglycoside resistance, presumably via active efflux. Aminoglycoside resistance caused by these two pumps could not be demonstrated in high-ionic-strength media, such as Luria broth or Mueller-Hinton broth. The EmrE-dependent efflux of ethidium bromide was confirmed by a continuous fluorescence assay.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antiporters/genetics , Bacterial Outer Membrane Proteins/genetics , Carrier Proteins/genetics , Drug Resistance, Bacterial/genetics , Membrane Proteins/genetics , Membrane Transport Proteins , Pseudomonas aeruginosa/genetics , Amino Acid Sequence , Aminoglycosides , Antiporters/drug effects , Bacterial Outer Membrane Proteins/drug effects , Carrier Proteins/drug effects , Escherichia coli Proteins , Membrane Proteins/drug effects , Microbial Sensitivity Tests , Plasmids , Pseudomonas aeruginosa/drug effects
20.
J Leukoc Biol ; 72(6): 1172-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12488499

ABSTRACT

The activation of human phagocytic leukocytes by immune complexes (IC) or opsonized microbes via their Fc and complement receptors has been well-described. The mechanisms involved in this process are complex and depend on the receptors involved. The biochemical events that lead to the destruction of invading organisms in turn display varying degrees of interdependence, but the controlling elements that lead to the ultimate killing of ingested organisms within phagosomes by lysosomal enzymes and reactive oxygen intermediates are still not completely understood. We have addressed these mechanisms by following and correlating the kinetics of responses by individual cells, using multiparameter flow cytometry. Using nonopsonized IC as stimuli, we document here the presence of a novel Ca(2)(+)/H(+) voltage-independent channel in human neutrophils, which helps to control their cytoplasmic pH.


Subject(s)
Antigen-Antibody Complex/physiology , Antiporters/physiology , Calcium-Binding Proteins/physiology , Cation Transport Proteins , Neutrophil Activation/drug effects , Antigen-Antibody Complex/pharmacology , Antiporters/drug effects , Calcium/metabolism , Calcium-Binding Proteins/drug effects , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Hydrogen-Ion Concentration , Kinetics , Membrane Potentials/physiology , Sodium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...