Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Front Pharmacol ; 14: 1293578, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38149052

RESUMEN

Introduction: Cystic fibrosis (CF) is caused by defective Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) proteins. CFTR controls chloride (Cl-) and bicarbonate (HCO3 -) transport into the Airway Surface Liquid (ASL). We investigated the impact of F508del-CFTR correction on HCO3 - secretion by studying transepithelial HCO3 - fluxes. Methods: HCO3 - secretion was measured by pH-stat technique in primary human respiratory epithelial cells from healthy subjects (WT) and people with CF (pwCF) carrying at least one F508del variant. Its changes after CFTR modulation by the triple combination VX445/661/770 and in the context of TNF-α+IL-17 induced inflammation were correlated to ASL pH and transcriptional levels of CFTR and other HCO3 - transporters of airway epithelia such as SLC26A4 (Pendrin), SLC26A9 and NBCe1. Results: CFTR-mediated HCO3 - secretion was not detected in F508del primary human respiratory epithelial cells. It was rescued up to ∼ 80% of the WT level by VX-445/661/770. In contrast, TNF-α+IL-17 normalized transepithelial HCO3 - transport and increased ASL pH. This was related to an increase in SLC26A4 and CFTR transcript levels. VX-445/661/770 induced an increase in pH only in the context of inflammation. Effects on HCO3 - transport were not different between F508del homozygous and F508del compound heterozygous CF airway epithelia. Conclusion: Our studies show that correction of F508del-CFTR HCO3 - is not sufficient to buffer acidic ASL and inflammation is a key regulator of HCO3 - secretion in CF airways. Prediction of the response to CFTR modulators by theratyping should take into account airway inflammation.

2.
Nat Commun ; 14(1): 4234, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37454114

RESUMEN

The chloroquine resistance transporter, PfCRT, of the human malaria parasite Plasmodium falciparum is sensitive to acidic pH. Consequently, PfCRT operates at 60% of its maximal drug transport activity at the pH of 5.2 of the digestive vacuole, a proteolytic organelle from which PfCRT expels drugs interfering with heme detoxification. Here we show by alanine-scanning mutagenesis that E207 is critical for pH sensing. The E207A mutation abrogates pH-sensitivity, while preserving drug substrate specificity. Substituting E207 with Asp or His, but not other amino acids, restores pH-sensitivity. Molecular dynamics simulations and kinetics analyses suggest an allosteric binding model in which PfCRT can accept both protons and chloroquine in a partial noncompetitive manner, with increased proton concentrations decreasing drug transport. Further simulations reveal that E207 relocates from a peripheral to an engaged location during the transport cycle, forming a salt bridge with residue K80. We propose that the ionized carboxyl group of E207 acts as a hydrogen acceptor, facilitating transport cycle progression, with pH sensing as a by-product.


Asunto(s)
Antimaláricos , Malaria Falciparum , Humanos , Antimaláricos/farmacología , Antimaláricos/química , Cloroquina/farmacología , Proteínas de Transporte de Membrana/metabolismo , Proteínas Protozoarias/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Concentración de Iones de Hidrógeno , Resistencia a Medicamentos/genética , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología
3.
JCI Insight ; 7(22)2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36219481

RESUMEN

The fluid covering the surface of airway epithelia represents a first barrier against pathogens. The chemical and physical properties of the airway surface fluid are controlled by the activity of ion channels and transporters. In cystic fibrosis (CF), loss of CFTR chloride channel function causes airway surface dehydration, bacterial infection, and inflammation. We investigated the effects of IL-17A plus TNF-α, 2 cytokines with relevant roles in CF and other chronic lung diseases. Transcriptome analysis revealed a profound change with upregulation of several genes involved in ion transport, antibacterial defense, and neutrophil recruitment. At the functional level, bronchial epithelia treated in vitro with the cytokine combination showed upregulation of ENaC channel, ATP12A proton pump, ADRB2 ß-adrenergic receptor, and SLC26A4 anion exchanger. The overall result of IL-17A/TNF-α treatment was hyperviscosity of the airway surface, as demonstrated by fluorescence recovery after photobleaching (FRAP) experiments. Importantly, stimulation with a ß-adrenergic agonist switched airway surface to a low-viscosity state in non-CF but not in CF epithelia. Our study suggests that CF lung disease is sustained by a vicious cycle in which epithelia cannot exit from the hyperviscous state, thus perpetuating the proinflammatory airway surface condition.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Humanos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Depuración Mucociliar , Interleucina-17/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Adrenérgicos/farmacología , Células Epiteliales/metabolismo , Fibrosis Quística/genética , Citocinas/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio
4.
JCI Insight ; 6(15)2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34166227

RESUMEN

Idiopathic nephrotic syndrome (INS) is characterized by proteinuria and renal sodium retention leading to edema. This sodium retention is usually attributed to epithelial sodium channel (ENaC) activation after plasma aldosterone increase. However, most nephrotic patients show normal aldosterone levels. Using a corticosteroid-clamped (CC) rat model of INS (CC-PAN), we showed that the observed electrogenic and amiloride-sensitive Na retention could not be attributed to ENaC. We then identified a truncated variant of acid-sensing ion channel 2b (ASIC2b) that induced sustained acid-stimulated sodium currents when coexpressed with ASIC2a. Interestingly, CC-PAN nephrotic ASIC2b-null rats did not develop sodium retention. We finally showed that the expression of the truncated ASIC2b in the kidney was dependent on the presence of albumin in the tubule lumen and activation of ERK in renal cells. Finally, the presence of ASIC2 mRNA was also detected in kidney biopsies from patients with INS but not in any of the patients with other renal diseases. We have therefore identified a variant of ASIC2b responsible for the renal Na retention in the pathological context of INS.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Riñón , Sistema de Señalización de MAP Quinasas , Síndrome Nefrótico , Canales de Sodio/metabolismo , Sodio , Albúminas/metabolismo , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Homeostasis , Riñón/metabolismo , Riñón/patología , Síndrome Nefrótico/sangre , Síndrome Nefrótico/metabolismo , Proteinuria/metabolismo , Ratas , Sodio/sangre , Sodio/metabolismo
5.
Int J Mol Sci ; 22(7)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33806154

RESUMEN

Knowledge on the mechanisms of acid and base secretion in airways has progressed recently. The aim of this review is to summarize the known mechanisms of airway surface liquid (ASL) pH regulation and their implication in lung diseases. Normal ASL is slightly acidic relative to the interstitium, and defects in ASL pH regulation are associated with various respiratory diseases, such as cystic fibrosis. Basolateral bicarbonate (HCO3-) entry occurs via the electrogenic, coupled transport of sodium (Na+) and HCO3-, and, together with carbonic anhydrase enzymatic activity, provides HCO3- for apical secretion. The latter mainly involves CFTR, the apical chloride/bicarbonate exchanger pendrin and paracellular transport. Proton (H+) secretion into ASL is crucial to maintain its relative acidity compared to the blood. This is enabled by H+ apical secretion, mainly involving H+/K+ ATPase and vacuolar H+-ATPase that carry H+ against the electrochemical potential gradient. Paracellular HCO3- transport, the direction of which depends on the ASL pH value, acts as an ASL protective buffering mechanism. How the transepithelial transport of H+ and HCO3- is coordinated to tightly regulate ASL pH remains poorly understood, and should be the focus of new studies.


Asunto(s)
Bicarbonatos/química , Anhidrasas Carbónicas/metabolismo , Epitelio/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Antiportadores/metabolismo , Antiportadores de Cloruro-Bicarbonato/metabolismo , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Ratones , Conejos , Transportadores de Sulfato/metabolismo , Tráquea/metabolismo
6.
Hum Mutat ; 41(4): 774-785, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31803959

RESUMEN

Pathological missense mutations in CLCNKB gene give a wide spectrum of clinical phenotypes in Bartter syndrome type III patients. Molecular analysis of the mutated ClC-Kb channels can be helpful to classify the mutations according to their functional alteration. We investigated the functional consequences of nine mutations in the CLCNKB gene causing Bartter syndrome. We first established that all tested mutations lead to decreased ClC-Kb currents. Combining electrophysiological and biochemical methods in Xenopus laevis oocytes and in MDCKII cells, we identified three classes of mutations. One class is characterized by altered channel trafficking. p.A210V, p.P216L, p.G424R, and p.G437R are totally or partially retained in the endoplasmic reticulum. p.S218N is characterized by reduced channel insertion at the plasma membrane and altered pH-sensitivity; thus, it falls in the second class of mutations. Finally, we found a novel class of functionally inactivated mutants normally present at the plasma membrane. Indeed, we found that p.A204T alters the pH-sensitivity, p.A254V abolishes the calcium-sensitivity. p.G219C and p.G465R are probably partially inactive at the plasma membrane. In conclusion, most pathogenic mutants accumulate partly or totally in intracellular compartments, but some mutants are normally present at the membrane surface and simultaneously show a large range of altered channel gating properties.


Asunto(s)
Síndrome de Bartter/genética , Sitios de Unión , Calcio/metabolismo , Canales de Cloruro/química , Canales de Cloruro/genética , Mutación , Multimerización de Proteína , Animales , Síndrome de Bartter/metabolismo , Línea Celular , Humanos , Oocitos/metabolismo , Unión Proteica , Transporte de Proteínas , Xenopus
8.
J Biol Chem ; 294(34): 12766-12778, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31285265

RESUMEN

The chloroquine resistance transporter PfCRT of the human malaria parasite Plasmodium falciparum confers resistance to the former first-line antimalarial drug chloroquine, and it modulates the responsiveness to a wide range of quinoline and quinoline-like compounds. PfCRT is post-translationally modified by phosphorylation, palmitoylation, and, possibly, ubiquitination. However, the impact of these post-translational modifications on P. falciparum biology and, in particular, the drug resistance-conferring activity of PfCRT has remained elusive. Here, we confirm phosphorylation at Ser-33 and Ser-411 of PfCRT of the chloroquine-resistant P. falciparum strain Dd2 and show that kinase inhibitors can sensitize drug responsiveness. Using CRISPR/Cas9 genome editing to generate genetically engineered PfCRT variants in the parasite, we further show that substituting Ser-33 with alanine reduced chloroquine and quinine resistance by ∼50% compared with the parental P. falciparum strain Dd2, whereas the phosphomimetic amino acid aspartic acid could fully and glutamic acid could partially reconstitute the level of chloroquine/quinine resistance. Transport studies conducted in the parasite and in PfCRT-expressing Xenopus laevis oocytes linked phosphomimetic substitution at Ser-33 to increased transport velocity. Our data are consistent with phosphorylation of Ser-33 relieving an autoinhibitory intramolecular interaction within PfCRT, leading to a stimulated drug transport activity. Our findings shed additional light on the function of PfCRT and suggest that chloroquine could be reevaluated as an antimalarial drug by targeting the kinase in P. falciparum that phosphorylates Ser-33 of PfCRT.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Serina/metabolismo , Antimaláricos/farmacología , Cloroquina/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Cinética , Pruebas de Sensibilidad Parasitaria , Fosforilación , Plasmodium falciparum/efectos de los fármacos , Proteínas Protozoarias/antagonistas & inhibidores
9.
Am J Physiol Renal Physiol ; 317(2): F435-F443, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31188029

RESUMEN

We have recently reported that type A intercalated cells of the collecting duct secrete Na+ by a mechanism coupling the basolateral type 1 Na+-K+-2Cl- cotransporter with apical type 2 H+-K+-ATPase (HKA2) functioning under its Na+/K+ exchange mode. The first aim of the present study was to evaluate whether this secretory pathway is a target of atrial natriuretic peptide (ANP). Despite hyperaldosteronemia, metabolic acidosis is not associated with Na+ retention. The second aim of the present study was to evaluate whether ANP-induced stimulation of Na+ secretion by type A intercalated cells might account for mineralocorticoid escape during metabolic acidosis. In Xenopus oocytes expressing HKA2, cGMP, the second messenger of ANP, increased the membrane expression, activity, and Na+-transporting rate of HKA2. Feeding mice with a NH4Cl-enriched diet increased urinary excretion of aldosterone and induced a transient Na+ retention that reversed within 3 days. At that time, expression of ANP mRNA in the collecting duct and urinary excretion of cGMP were increased. Reversion of Na+ retention was prevented by treatment with an inhibitor of ANP receptors and was absent in HKA2-null mice. In conclusion, paracrine stimulation of HKA2 by ANP is responsible for the escape of the Na+-retaining effect of aldosterone during metabolic acidosis.


Asunto(s)
Equilibrio Ácido-Base , Acidosis/enzimología , Factor Natriurético Atrial/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Túbulos Renales Colectores/enzimología , Sodio/orina , Acidosis/genética , Acidosis/fisiopatología , Acidosis/orina , Adaptación Fisiológica , Aldosterona/orina , Animales , GMP Cíclico/orina , Femenino , ATPasa Intercambiadora de Hidrógeno-Potásio/deficiencia , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Concentración de Iones de Hidrógeno , Ratones Endogámicos C57BL , Ratones Noqueados , Comunicación Paracrina , Ratas , Transducción de Señal , Xenopus laevis
10.
Sci Rep ; 9(1): 6516, 2019 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-31019198

RESUMEN

Cystic fibrosis (CF) is caused by defective Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) protein. Morbidity is mainly due to early airway infection. We hypothesized that S. aureus clearance during the first hours of infection was impaired in CF human Airway Surface Liquid (ASL) because of a lowered pH. The ASL pH of human bronchial epithelial cell lines and primary respiratory cells from healthy controls (WT) and patients with CF was measured with a pH microelectrode. The antimicrobial capacity of airway cells was studied after S. aureus apical infection by counting surviving bacteria. ASL was significantly more acidic in CF than in WT respiratory cells. This was consistent with a defect in bicarbonate secretion involving CFTR and SLC26A4 (pendrin) and a persistent proton secretion by ATP12A. ASL demonstrated a defect in S. aureus clearance which was improved by pH normalization. Pendrin inhibition in WT airways recapitulated the CF airway defect and increased S. aureus proliferation. ATP12A inhibition by ouabain decreased bacterial proliferation. Antimicrobial peptides LL-37 and hBD1 demonstrated a pH-dependent activity. Normalizing ASL pH might improve innate airway defense in newborns with CF during onset of S. aureus infection. Pendrin activation and ATP12A inhibition could represent novel therapeutic strategies to normalize pH in CF airways.


Asunto(s)
Bronquios/citología , Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Mucosa Respiratoria/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Bicarbonatos/química , Bicarbonatos/metabolismo , Línea Celular , Células Cultivadas , Niño , Preescolar , Fibrosis Quística/genética , Fibrosis Quística/microbiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Lactante , Recién Nacido , Mucosa Respiratoria/química , Mucosa Respiratoria/microbiología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/fisiología , Transportadores de Sulfato/metabolismo , Catelicidinas
11.
Compr Physiol ; 9(1): 301-342, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30549019

RESUMEN

The many mechanisms governing NaCl absorption in the diverse parts of the renal tubule have been largely elucidated, although some of them, as neutral NaCl absorption across the cortical collecting duct or regulation through with-no-lysine (WNK) kinases have emerged only recently. Chloride channels, which are important players in these processes, at least in the distal nephron, are the focus of this review. Over the last 20-year period, experimental studies using molecular, electrophysiological, and physiological/functional approaches have deepened and renewed our views on chloride channels and their role in renal function. Two chloride channels of the ClC family, named as ClC-Ka and ClC-Kb in humans and ClC-K1 and ClC-K2 in other mammals, are preponderant and play complementary roles: ClC-K1/Ka is mainly involved in the building of the interstitial cortico-medullary concentration gradient, while ClC-K2/Kb participates in NaCl absorption in the thick ascending limb, distal convoluted tubule and the intercalated cells of the collecting duct. The two ClC-Ks might also be involved indirectly in proton secretion by type A intercalated cells. Other chloride channels in the kidneys include CFTR, TMEM16A, and probably volume-regulated LRRC8 chloride channels, whose function and molecular identity have not as yet been established. © 2019 American Physiological Society. Compr Physiol 9:301-342, 2019.


Asunto(s)
Canales de Cloruro/metabolismo , Riñón/metabolismo , Cloruro de Sodio/metabolismo , Animales , Canales de Cloruro/química , Canales de Cloruro/genética , Humanos , Riñón/fisiología , Reabsorción Renal
12.
JCI Insight ; 3(20)2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30333310

RESUMEN

Proton secretion mediated by ATP12A protein on the surface of the airway epithelium may contribute to cystic fibrosis (CF) lung disease by favoring bacterial infection and airway obstruction. We studied ATP12A in fresh bronchial samples and in cultured epithelial cells. In vivo, ATP12A expression was found almost exclusively at the apical side of nonciliated cells of airway epithelium and in submucosal glands, with much higher expression in CF samples. This could be due to bacterial infection and inflammation, since treating cultured cells with bacterial supernatants or with IL-4 (a cytokine that induces goblet cell hyperplasia) increased the expression of ATP12A in nonciliated cells. This observation was associated with upregulation and translocation of ATP1B1 protein from the basal to apical epithelial side, where it colocalizes with ATP12A. ATP12A function was evaluated by measuring the pH of the apical fluid in cultured epithelia. Under resting conditions, CF epithelia showed more acidic values. This abnormality was minimized by inhibiting ATP12A with ouabain. Following treatment with IL-4, ATP12A function was markedly increased, as indicated by strong acidification occurring under bicarbonate-free conditions. Our study reveals potentially novel aspects of ATP12A and remarks its importance as a possible therapeutic target in CF and other respiratory diseases.


Asunto(s)
Bronquios/patología , Fibrosis Quística/patología , Células Caliciformes/patología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Animales , Bronquios/citología , Bronquios/inmunología , Membrana Celular/metabolismo , Células Cultivadas , Colon/citología , Colon/metabolismo , Fibrosis Quística/inmunología , Fibrosis Quística/cirugía , Células Caliciformes/inmunología , Células Caliciformes/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Humanos , Concentración de Iones de Hidrógeno , Interleucina-4/inmunología , Interleucina-4/metabolismo , Ratones , Ratones Noqueados , Ouabaína/farmacología , Permeabilidad , Potasio/metabolismo , Cultivo Primario de Células , Inhibidores de la Bomba de Protones/farmacología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
13.
Sci Rep ; 7(1): 7249, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28775266

RESUMEN

The human ClC-Kb channel plays a key role in exporting chloride ions from the cytosol and is known to be involved in Bartter syndrome type 3 when its permeation capacity is decreased. The ClC-Kb channel has been recently proposed as a potential therapeutic target to treat hypertension. In order to gain new insights into the sequence-structure-function relationships of this channel, to investigate possible impacts of amino-acid substitutions, and to design novel inhibitors, we first built a structural model of the human ClC-Kb channel using comparative modeling strategies. We combined in silico and in vitro techniques to analyze amino acids involved in the chloride ion pathway as well as to rationalize the possible role of several clinically observed mutations leading to the Bartter syndrome type 3. Virtual screening and drug repositioning computations were then carried out. We identified six novel molecules, including 2 approved drugs, diflusinal and loperamide, with Kd values in the low micromolar range, that block the human ClC-Kb channel and that could be used as starting point to design novel chemical probes for this potential therapeutic target.


Asunto(s)
Canales de Cloruro/química , Modelos Moleculares , Relación Estructura-Actividad Cuantitativa , Secuencia de Aminoácidos , Animales , Bovinos , Canales de Cloruro/antagonistas & inhibidores , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Cloruros/química , Cloruros/metabolismo , Susceptibilidad a Enfermedades , Evaluación Preclínica de Medicamentos , Humanos , Activación del Canal Iónico , Potenciales de la Membrana , Estructura Molecular , Mutación , Conformación Proteica
14.
J Biol Chem ; 292(39): 16109-16121, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28768767

RESUMEN

The chloroquine resistance transporter of the human malaria parasite Plasmodium falciparum, PfCRT, is an important determinant of resistance to several quinoline and quinoline-like antimalarial drugs. PfCRT also plays an essential role in the physiology of the parasite during development inside erythrocytes. However, the function of this transporter besides its role in drug resistance is still unclear. Using electrophysiological and flux experiments conducted on PfCRT-expressing Xenopus laevis oocytes, we show here that both wild-type PfCRT and a PfCRT variant associated with chloroquine resistance transport both ferrous and ferric iron, albeit with different kinetics. In particular, we found that the ability to transport ferrous iron is reduced by the specific polymorphisms acquired by the PfCRT variant as a result of chloroquine selection. We further show that iron and chloroquine transport via PfCRT is electrogenic. If these findings in the Xenopus model extend to P. falciparum in vivo, our data suggest that PfCRT might play a role in iron homeostasis, which is essential for the parasite's development in erythrocytes.


Asunto(s)
Antimaláricos/metabolismo , Cloroquina/metabolismo , Hierro/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Sustitución de Aminoácidos , Animales , Transporte Biológico , Hierro/química , Cinética , Proteínas de Transporte de Membrana/genética , Mutación , Oocitos/metabolismo , Oxidación-Reducción , Técnicas de Placa-Clamp , Proteínas Protozoarias/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
15.
J Mol Biol ; 428(14): 2898-915, 2016 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-27241308

RESUMEN

Deletion of Phe508 in the nucleotide binding domain (∆F508-NBD1) of the cystic fibrosis transmembrane regulator (CFTR; a cyclic AMP-regulated chloride channel) is the most frequent mutation associated with cystic fibrosis. This mutation affects the maturation and gating of CFTR protein. The search for new high-affinity ligands of CFTR acting as dual modulators (correctors/activators) presents a major challenge in the pharmacology of cystic fibrosis. Snake venoms are a rich source of natural multifunctional proteins, potential binders of ion channels. In this study, we identified the CB subunit of crotoxin from Crotalus durissus terrificus as a new ligand and allosteric modulator of CFTR. We showed that CB interacts with NBD1 of both wild type and ∆F508CFTR and increases their chloride channel currents. The potentiating effect of CB on CFTR activity was demonstrated using electrophysiological techniques in Xenopus laevis oocytes, in CFTR-HeLa cells, and ex vivo in mouse colon tissue. The correcting effect of CB was shown by functional rescue of CFTR activity after 24-h ΔF508CFTR treatments with CB. Moreover, the presence of fully glycosylated CFTR was observed. Molecular docking allowed us to propose a model of the complex involving of the ABCß and F1-like ATP-binding subdomains of ΔF508-NBD1. Hydrogen-deuterium exchange analysis confirmed stabilization in these regions, also showing allosteric stabilization in two other distal regions. Surface plasmon resonance competition studies showed that CB disrupts the ∆F508CFTR-cytokeratin 8 complex, allowing for the escape of ∆F508CFTR from degradation. Therefore CB, as a dual modulator of ΔF508CFTR, constitutes a template for the development of new anti-CF agents.


Asunto(s)
Canales de Cloruro/genética , Crotalus/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Fosfolipasas A2/genética , Venenos de Serpiente/genética , Animales , Línea Celular Tumoral , AMP Cíclico/genética , Femenino , Células HeLa , Humanos , Activación del Canal Iónico/genética , Cinética , Masculino , Ratones , Simulación del Acoplamiento Molecular/métodos , Mutación/genética , Oocitos/metabolismo , Unión Proteica/genética , Eliminación de Secuencia/genética , Xenopus laevis/genética
16.
Hum Mutat ; 34(10): 1404-14, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24272871

RESUMEN

Diffuse bronchiectasis is a common problem in respiratory clinics. We hypothesized that mutations in the solute carrier 26A9 (SLC26A9) gene, encoding for a chloride (Cl(-)) transporter mainly expressed in lungs, may lead to defects in mucociliary clearance. We describe two missense variants in the SLC26A9 gene in heterozygote patients presenting with diffuse idiopathic bronchiectasis : p.Arg575Trp, identified in a patient also heterozygote for p.Phe508del in the CFTR gene; and p.Val486Ile. Expression of both mutants in Xenopus laevis oocytes abolished SLC26A9-mediated Cl(-) conductance without decreasing protein membrane expression. Coexpression of CFTR with SLC26A9-p.Val486Ile resulted in a significant increase in the Cl(-) current induced by PKA stimulation, similar to that obtained in oocytes expressing CFTR and SLC26A9-WT. In contrast, coexpression of CFTR with SLC26A9-p.Arg575Trp inhibited SLC26A9-enhanced CFTR activation upon PKA. Further structure-function analyses led us to propose a site encompassing Arg575 in the SLC26A9-STAS domain for CFTR-SLC26A9 interaction. We hypothesize that SLC26A9-p.Arg575Trp prevented SLC26A9-mediated functional activation of CFTR by altering SLC26A9-CFTR interaction. Although we cannot confirm that these mutations by themselves are deleterious, we propose that they trigger the pathogenic role of a single CFTR mutation and provide insight into a novel mechanism of Cl(-) transport alteration across the respiratory mucosa, based on functional inhibition of CFTR.


Asunto(s)
Antiportadores/genética , Enfermedades Pulmonares/diagnóstico , Enfermedades Pulmonares/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antiportadores/química , Antiportadores/metabolismo , Estudios de Casos y Controles , Niño , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Exones , Femenino , Expresión Génica , Humanos , Enfermedades Pulmonares/patología , Masculino , Persona de Mediana Edad , Mutación , Oocitos/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fenotipo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transportadores de Sulfato , Tomografía Computarizada por Rayos X , Xenopus laevis , Adulto Joven
17.
J Inherit Metab Dis ; 36(1): 103-12, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22644605

RESUMEN

Intellectual disability coupled with epilepsy are clinical hallmarks of the creatine (Cr) transporter deficiency syndrome resulting from mutations in the SLC6A8 gene. So far characterization of pathogenic mutations of SLC6A8 has been limited to Cr uptake. The aim of our study was to characterize the electrogenic and pharmacological properties of non truncating SLC6A8 mutations identified in patients presenting variable clinical severity. Electrophysiological and pharmacological properties of four mutants (including two novel ones) were studied in X. laevis oocyte expression system. Creatine uptake was assessed with [(14)C]-Cr in X. laevis and patients' fibroblasts. Subcellular localization was determined by immunofluorescence and western blot. All mutants were properly targeted to the plasma membrane in both systems. Mutations led to the complete loss of both electrogenic and transport activities in X. laevis and Cr uptake in patients' fibroblasts. Among the Cr analogs tested, guanidinopropionate induced an electrogenic activity with the normal SLC6A8 transporter similar to creatine whereas a phosphocreatine derivative, PCr-Mg-CPLX, resulted in partial activity. SLC6A8 mutants displayed no electrogenic activity with all Cr analogs tested in X. laevis oocytes. Although the mutations altered various domains of SLC6A8 Cr uptake and electrogenic properties were completely inhibited and could not be dissociated. Besides the metabolic functions of Cr, the loss of SLC6A8 electrogenic activity, demonstrated here for the first time, may also play a role in the altered brain functions of the patients.


Asunto(s)
Proteínas de Transporte de Membrana/genética , Mutación , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/deficiencia , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética , Animales , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Niño , Preescolar , Creatina/genética , Creatina/metabolismo , Fenómenos Electrofisiológicos , Fibroblastos/metabolismo , Genotipo , Humanos , Masculino , Proteínas de Transporte de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oocitos/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo
18.
Int J Biochem Cell Biol ; 44(6): 1009-18, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22481026

RESUMEN

Prostaglandins, the products of arachidonic acid release and oxidation by phospholipase A(2) and cyclooxygenases (COX) 1 and 2 respectively, are known as important inflammation mediators. However, their diversity in structure, properties and cell specificity make their physiological function difficult to define. In the lung, the prostaglandin D(2) (PGD(2)) metabolite 15d-PGJ(2) is known to modulate the properties of a large number of intracellular compounds, leading to both pro- and anti-inflammatory effects. In the lung, the serous sub-mucosal glands, that strongly express CFTR (cystic fibrosis transmembrane conductance regulator), play an important role in the defence against inflammation, and their derivatives Calu-3 cells are largely used in in vitro experiments. The present study was undertaken to determine whether the PGD synthase-PGD(2)-15d-PGJ(2) pathway is active in Calu-3 cells, and whether its activity requires a functional CFTR. Both cellular and released PGD(2) and 15d-PGJ(2) were measured in cells treated with CFTR inhibitors and stimulated or not with inflammatory IL-1ß. Pretreatment with either CFTR(inh172) or GlyH101 inhibitors decreased the basal cell content of both prostaglandins, and so did acute stimulation with IL-1ß, but the latter was dramatically reversed in CFTR(inh172)-treated cells. CFTR(inh172) also altered the release of inflammation mediators PGE(2) and IL-8, and this effect was blunted by exogenous 15d-PGJ(2). CFTR(inh172)-induced modulation of 15d-PGJ(2) cellular content was not detected in CFTR-silenced Calu-3 cells, but it was reproduced in pulmonary CFBE41o-cells, which express F508del-CFTR. These results show that cellular 15d-PGJ(2) production, which controls PGE(2) and IL-8 release, is disturbed by CFTR dysfunction. In Calu-3 cells, 15d-PGJ(2) production resulted from COX-2-regulated COX-1 activation, while CFTR(inh172)-induced alteration of 15d-PGJ(2) synthesis involved both decreased expression of PGD synthase and disturbed relationships between both COXs. CFTR-mediated regulation of PGD synthase-PGD(2)-15d-PGJ(2) pathway and cellular 15d-PGJ(2) effects may involve a large number of molecular reactive pathways. Their exploration should help understand the development of CF inflammation and might bring new perspectives in its treatment.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Prostaglandina D2/análogos & derivados , Western Blotting , Línea Celular , Humanos , Interleucina-1beta/antagonistas & inhibidores , Pulmón/citología , Pulmón/metabolismo , Prostaglandina D2/farmacología
19.
PLoS One ; 7(4): e34879, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22514683

RESUMEN

BACKGROUND: A growing number of proteins, including ion transporters, have been shown to interact with Cystic Fibrosis Transmembrane conductance Regulator (CFTR). CFTR is an epithelial chloride channel that is involved in Cystic Fibrosis (CF) when mutated; thus a better knowledge of its functional interactome may help to understand the pathophysiology of this complex disease. In the present study, we investigated if CFTR and the sodium-phosphate co-transporter type 2a (NPT2a) functionally interact after heterologous expression of both proteins in Xenopus laevis oocytes. METHODOLOGY/FINDINGS: NPT2a was expressed alone or in combination with CFTR in X. laevis oocytes. Using the two-electrode voltage-clamp technique, the inorganic phosphate-induced current (IPi) was measured and taken as an index of NPT2a activity. The maximal IPi for NPT2a substrates was reduced when CFTR was co-expressed with NPT2a, suggesting a decrease in its expression at the oolemna. This was consistent with Western blot analysis showing reduced NPT2a plasma membrane expression in oocytes co-expressing both proteins, whereas NPT2a protein level in total cell lysate was the same in NPT2a- and NPT2a+CFTR-oocytes. In NPT2a+CFTR- but not in NPT2a-oocytes, IPi and NPT2a surface expression were increased upon PKA stimulation, whereas stimulation of Exchange Protein directly Activated by cAMP (EPAC) had no effect. When NPT2a-oocytes were injected with NEG2, a short amino-acid sequence from the CFTR regulatory domain that regulates PKA-dependent CFTR trafficking to the plasma membrane, IPi values and NPT2a membrane expression were diminished, and could be enhanced by PKA stimulation, thereby mimicking the effects of CFTR co-expression. CONCLUSION/PERSPECTIVES: We conclude that when both CFTR and NPT2a are expressed in X. laevis oocytes, CFTR confers to NPT2a a cAMPi-dependent trafficking to the membrane. This functional interaction raises the hypothesis that CFTR may play a role in phosphate homeostasis.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Oocitos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animales , Femenino , Unión Proteica
20.
PLoS One ; 7(4): e34764, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22506049

RESUMEN

BACKGROUND: The sodium-hydrogen exchanger regulatory factor 1 (NHERF1) binds to the main renal phosphate transporter NPT2a and to the parathyroid hormone (PTH) receptor. We have recently identified mutations in NHERF1 that decrease renal phosphate reabsorption by increasing PTH-induced cAMP production in the renal proximal tubule. METHODS: We compared relevant parameters of phosphate homeostasis in a patient with a previously undescribed mutation in NHERF1 and in control subjects. We expressed the mutant NHERF1 protein in Xenopus Oocytes and in cultured cells to study its effects on phosphate transport and PTH-induced cAMP production. RESULTS: We identified in a patient with inappropriate renal phosphate reabsorption a previously unidentified mutation (E68A) located in the PDZ1 domain of NHERF1.We report the consequences of this mutation on NHERF1 function. E68A mutation did not modify cAMP production in the patient. PTH-induced cAMP synthesis and PKC activity were not altered by E68A mutation in renal cells in culture. In contrast to wild-type NHERF1, expression of the E68A mutant in Xenopus oocytes and in human cells failed to increase phosphate transport. Pull down experiments showed that E68A mutant did not interact with NPT2a, which robustly interacted with wild type NHERF1 and previously identified mutants. Biotinylation studies revealed that E68A mutant was unable to increase cell surface expression of NPT2a. CONCLUSIONS: Our results indicate that the PDZ1 domain is critical for NHERF1-NPT2a interaction in humans and for the control of NPT2a expression at the plasma membrane. Thus we have identified a new mechanism of renal phosphate loss and shown that different mutations in NHERF1 can alter renal phosphate reabsorption via distinct mechanisms.


Asunto(s)
Mutación , Hormona Paratiroidea/metabolismo , Proteínas de Transporte de Fosfato/genética , Fosfoproteínas/genética , Intercambiadores de Sodio-Hidrógeno/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Anciano , Animales , Línea Celular Tumoral , Células Cultivadas , AMP Cíclico/metabolismo , Células HeLa , Humanos , Túbulos Renales Proximales/metabolismo , Oocitos/metabolismo , Zarigüeyas , Proteínas de Transporte de Fosfato/biosíntesis , Fosfatos/metabolismo , Fosfoproteínas/metabolismo , Proteína Quinasa C/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...