Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Am J Physiol Cell Physiol ; 323(2): C385-C399, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35759442

RESUMEN

The thiazide-sensitive Na+-Cl- cotransporter (NCC) is the major pathway for salt reabsorption in the mammalian distal convoluted tubule, and the inhibition of its function with thiazides is widely used for the treatment of arterial hypertension. In mammals and teleosts, NCC is present as one ortholog that is mainly expressed in the kidney. One exception, however, is the eel, which has two genes encoding NCC. The eNCCα is located in the kidney and eNCCß, which is present in the apical membrane of the rectum. Interestingly, the European eNCCß functions as a Na+-Cl- cotransporter that is nevertheless resistant to thiazides and is not activated by low-chloride hypotonic stress. However, in the Japanese eel rectal sac, a thiazide-sensitive NaCl transport mechanism has been described. The protein sequences between eNCCß and jNCCß are 98% identical. Here, by site-directed mutagenesis, we transformed eNCCß into jNCCß. Our data showed that jNCCß, similar to eNCCß, is resistant to thiazides. In addition, both NCCß proteins have high transport capacity with respect to their renal NCC orthologs and, in contrast to known NCCs, exhibit electrogenic properties that are reduced when residue I172 is substituted by A, G, or M. This is considered a key residue for the chloride ion-binding sites of NKCC and KCC. We conclude that NCCß proteins are not sensitive to thiazides and have electrogenic properties dependent on Cl-, and site I172 is important for the function of NCCß.


Asunto(s)
Cloruros , Inhibidores de los Simportadores del Cloruro de Sodio , Animales , Cloruros/metabolismo , Anguilas/metabolismo , Mamíferos/metabolismo , Cloruro de Sodio , Inhibidores de los Simportadores del Cloruro de Sodio/metabolismo , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Simportadores del Cloruro de Sodio/genética , Simportadores del Cloruro de Sodio/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Tiazidas/farmacología
2.
Curr Top Membr ; 83: 177-204, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31196605

RESUMEN

The thiazide-sensitive Na+-Cl- cotransporter (NCC) is the major pathway for salt reabsorption in the distal convoluted tubule, serves as a receptor for thiazide-type diuretics, and is involved in inherited diseases associated with abnormal blood pressure. The functional and structural characterization of NCC from different species has led us to gain insights into the structure-function relationships of the cotransporter. Here we present an overview of different studies that had described these properties. Additionally, we report the cloning and characterization of the NCC from the spiny dogfish (Squalus acanthias) kidney (sNCC). The purpose of the present study was to determine the main functional, pharmacological and regulatory properties of sNCC to make a direct comparison with other NCC orthologous. The sNCC cRNA encodes a 1033 amino acid membrane protein, when expressed in Xenopus oocytes, functions as a thiazide-sensitive Na-Cl cotransporter with NCC regulation and thiazide-inhibition properties similar to mammals, rather than to teleosts. However, the Km values for ion transport kinetics are significantly higher than those observed in the mammal species. In summary, we present a review on NCC structure-function relationships with the addition of the sNCC information in order to enrich the NCC cotransporter knowledge.


Asunto(s)
Riñón/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/química , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Síndrome de Gitelman/genética , Humanos , Mutación , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Relación Estructura-Actividad
3.
J Am Soc Nephrol ; 29(7): 1838-1848, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29848507

RESUMEN

Background Hypercalciuria can result from activation of the basolateral calcium-sensing receptor (CaSR), which in the thick ascending limb of Henle's loop controls Ca2+ excretion and NaCl reabsorption in response to extracellular Ca2+ However, the function of CaSR in the regulation of NaCl reabsorption in the distal convoluted tubule (DCT) is unknown. We hypothesized that CaSR in this location is involved in activating the thiazide-sensitive NaCl cotransporter (NCC) to prevent NaCl loss.Methods We used a combination of in vitro and in vivo models to examine the effects of CaSR on NCC activity. Because the KLHL3-WNK4-SPAK pathway is involved in regulating NaCl reabsorption in the DCT, we assessed the involvement of this pathway as well.Results Thiazide-sensitive 22Na+ uptake assays in Xenopus laevis oocytes revealed that NCC activity increased in a WNK4-dependent manner upon activation of CaSR with Gd3+ In HEK293 cells, treatment with the calcimimetic R-568 stimulated SPAK phosphorylation only in the presence of WNK4. The WNK4 inhibitor WNK463 also prevented this effect. Furthermore, CaSR activation in HEK293 cells led to phosphorylation of KLHL3 and WNK4 and increased WNK4 abundance and activity. Finally, acute oral administration of R-568 in mice led to the phosphorylation of NCC.Conclusions Activation of CaSR can increase NCC activity via the WNK4-SPAK pathway. It is possible that activation of CaSR by Ca2+ in the apical membrane of the DCT increases NaCl reabsorption by NCC, with the consequent, well known decrease of Ca2+ reabsorption, further promoting hypercalciuria.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Sensibles al Calcio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sodio/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Activación Enzimática/genética , Células HEK293 , Humanos , Imidazoles/farmacología , Masculino , Ratones , Proteínas de Microfilamentos , Oocitos , Fenetilaminas/farmacología , Fosforilación/efectos de los fármacos , Propilaminas/farmacología , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Pirrolidinas/farmacología , Receptores Sensibles al Calcio/genética , Transducción de Señal , Miembro 1 de la Familia de Transportadores de Soluto 12/antagonistas & inhibidores , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Transfección , Proteínas de Xenopus/metabolismo , Xenopus laevis
4.
J Biol Chem ; 291(43): 22472-22481, 2016 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-27587391

RESUMEN

The thiazide-sensitive Na-Cl cotransporter (NCC) is the major pathway for salt reabsorption in the mammalian distal convoluted tubule. NCC plays a key role in the regulation of blood pressure. Its inhibition with thiazides constitutes the primary baseline therapy for arterial hypertension. However, the thiazide-binding site in NCC is unknown. Mammals have only one gene encoding for NCC. The eel, however, contains a duplicate gene. NCCα is an ortholog of mammalian NCC and is expressed in the kidney. NCCß is present in the apical membrane of the rectum. Here we cloned and functionally characterized NCCß from the European eel. The cRNA encodes a 1043-amino acid membrane protein that, when expressed in Xenopus oocytes, functions as an Na-Cl cotransporter with two major characteristics, making it different from other known NCCs. First, eel NCCß is resistant to thiazides. Single-point mutagenesis supports that the absence of thiazide inhibition is, at least in part, due to the substitution of a conserved serine for a cysteine at position 379. Second, NCCß is not activated by low-chloride hypotonic stress, although the unique Ste20-related proline alanine-rich kinase (SPAK) binding site in the amino-terminal domain is conserved. Thus, NCCß exhibits significant functional differences from NCCs that could be helpful in defining several aspects of the structure-function relationship of this important cotransporter.


Asunto(s)
Resistencia a Medicamentos/efectos de los fármacos , Anguilas/metabolismo , Proteínas de Peces/metabolismo , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Simportadores del Cloruro de Sodio/metabolismo , Animales , Anguilas/genética , Proteínas de Peces/genética , Humanos , Oocitos , Ratas , Simportadores del Cloruro de Sodio/genética , Xenopus laevis
5.
Am J Physiol Cell Physiol ; 311(5): C720-C734, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27488665

RESUMEN

SMCTs move several important fuel molecules that are involved in lipid, carbohydrate, and amino acid metabolism, but their regulation has been poorly studied. Insulin controls the translocation of several solutes that are involved in energetic cellular metabolism, including glucose. We studied the effect of insulin on the function of human SMCT1 expressed in Xenopus oocytes. The addition of insulin reduced α-keto-isocaproate (KIC)-dependent 22Na+ uptake by 29%. Consistent with this result, the coinjection of SMCT1 with SGK1 cRNA decreased the KIC-dependent 22Na+ uptake by 34%. The reduction of SMCT1 activity by SGK1 depends on its kinase activity, and it was observed that the coinjection of SMCT1 with S442D-SGK1 (a constitutively active mutant) decreased the KIC-dependent 22Na+ uptake by 50%. In contrast, an SMCT1 coinjection with K127M-SGK1 (an inactive mutant) had no effect on the KIC-dependent Na+ uptake. The decreasing SMCT1 function by insulin or SGK1 was corroborated by measuring [1-14C]acetate uptake and the electric currents of SMCT1-injected oocytes. Previously, we found that SMCT2/Slc5a12-mRNA, but not SMCT1/Slc5a8-mRNA, is present in zebrafish pancreas (by in situ hybridization); however, SLC5a8 gene silencing was associated with the development of human pancreatic cancer. We confirmed that the mRNA and protein of both transporters were present in rat pancreas using RT-PCR with specific primers, Western blot analysis, and immunohistochemistry. Additionally, significant propionate-dependent 22Na+ uptake occurred in pancreatic islets and was reduced by insulin treatment. Our data indicate that human SMCT1 is regulated by insulin and SGK1 and that both SMCTs are present in the mammalian pancreas.


Asunto(s)
Proteínas Inmediatas-Precoces/metabolismo , Insulina/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Sodio/metabolismo , Animales , ADN Complementario/metabolismo , Humanos , Masculino , Oocitos/metabolismo , Páncreas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Xenopus laevis/metabolismo , Pez Cebra/metabolismo
6.
J Am Soc Nephrol ; 26(8): 1781-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25542968

RESUMEN

It is widely recognized that the phenotype of familial hyperkalemic hypertension is mainly a consequence of increased activity of the renal Na(+)-Cl(-) cotransporter (NCC) because of altered regulation by with no-lysine-kinase 1 (WNK1) or WNK4. The effect of WNK4 on NCC, however, has been controversial because both inhibition and activation have been reported. It has been recently shown that the long isoform of WNK1 (L-WNK1) is a chloride-sensitive kinase activated by a low Cl(-) concentration. Therefore, we hypothesized that WNK4 effects on NCC could be modulated by intracellular chloride concentration ([Cl(-)]i), and we tested this hypothesis in oocytes injected with NCC cRNA with or without WNK4 cRNA. At baseline in oocytes, [Cl(-)]i was near 50 mM, autophosphorylation of WNK4 was undetectable, and NCC activity was either decreased or unaffected by WNK4. A reduction of [Cl(-)]i, either by low chloride hypotonic stress or coinjection of oocytes with the solute carrier family 26 (anion exchanger)-member 9 (SLC26A9) cRNA, promoted WNK4 autophosphorylation and increased NCC-dependent Na(+) transport in a WNK4-dependent manner. Substitution of the leucine with phenylalanine at residue 322 of WNK4, homologous to the chloride-binding pocket in L-WNK1, converted WNK4 into a constitutively autophosphorylated kinase that activated NCC, even without chloride depletion. Elimination of the catalytic activity (D321A or D321K-K186D) or the autophosphorylation site (S335A) in mutant WNK4-L322F abrogated the positive effect on NCC. These observations suggest that WNK4 can exert differential effects on NCC, depending on the intracellular chloride concentration.


Asunto(s)
Cloruros/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Humanos , Ratones , Xenopus laevis
7.
Toxicology ; 502: 153726, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38191021

RESUMEN

Heavy metals are found naturally in our environment and have many uses and applications in daily life. However, high concentrations of metals may be a result of pollution due to industrialization. In particular, cadmium (Cd), a white metal abundantly distributed in the terrestrial crust, is found in mines together with zinc, which accumulates after volcanic eruption or is found naturally in the sea and earth. High levels of Cd have been associated with disease. In the human body, Cd accumulates in two ways: via inhalation or consumption, mainly of plants or fish contaminated with high concentrations. Several international organizations have been working to establish the limit values of heavy metals in food, water, and the environment to avoid their toxic effects. Increased Cd levels may induce kidney, liver, or neurological diseases. Cd mainly accumulates in the kidney, causing renal disease in people exposed to moderate to high levels, which leads to the development of end-stage chronic kidney disease or death. The aim of this review is to provide an overview of Cd-induced nephrotoxicity, the mechanisms of Cd damage, and the current treatments used to reduce the toxic effects of Cd exposure.


Asunto(s)
Cadmio , Metales Pesados , Humanos , Animales , Cadmio/toxicidad , Metales Pesados/toxicidad , Riñón , Hígado , Zinc/farmacología
8.
PLoS One ; 19(4): e0302436, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38662786

RESUMEN

Severe cases of COVID-19 are characterized by development of acute respiratory distress syndrome (ARDS). Water accumulation in the lungs is thought to occur as consequence of an exaggerated inflammatory response. A possible mechanism could involve decreased activity of the epithelial Na+ channel, ENaC, expressed in type II pneumocytes. Reduced transepithelial Na+ reabsorption could contribute to lung edema due to reduced alveolar fluid clearance. This hypothesis is based on the observation of the presence of a novel furin cleavage site in the S protein of SARS-CoV-2 that is identical to the furin cleavage site present in the alpha subunit of ENaC. Proteolytic processing of αENaC by furin-like proteases is essential for channel activity. Thus, competition between S protein and αENaC for furin-mediated cleavage in SARS-CoV-2-infected cells may negatively affect channel activity. Here we present experimental evidence showing that coexpression of the S protein with ENaC in a cellular model reduces channel activity. In addition, we show that bidirectional competition for cleavage by furin-like proteases occurs between 〈ENaC and S protein. In transgenic mice sensitive to lethal SARS-CoV-2, however, a significant decrease in gamma ENaC expression was not observed by immunostaining of lungs infected as shown by SARS-CoV2 nucleoprotein staining.


Asunto(s)
COVID-19 , Canales Epiteliales de Sodio , Furina , Ratones Transgénicos , Proteolisis , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Canales Epiteliales de Sodio/metabolismo , Animales , Humanos , Ratones , Furina/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo , COVID-19/metabolismo , COVID-19/virología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , Pulmón/metabolismo , Pulmón/virología , Pulmón/patología , Células HEK293
9.
Am J Physiol Cell Physiol ; 302(8): C1083-95, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22159080

RESUMEN

Marine fish drink seawater and eliminate excess salt by active salt transport across gill and gut epithelia. Euryhaline pufferfish (Takifugu obscurus, mefugu) forms a CaCO(3) precipitate on the luminal gut surface after transitioning to seawater. NBCe1 (Slc4a4) at the basolateral membrane of intestinal epithelial cell plays a major role in transepithelial intestinal HCO(3)(-) secretion and is critical for mefugu acclimation to seawater. We assayed fugu-NBCe1 (fNBCe1) activity in the Xenopus oocyte expression system. Similar to NBCe1 found in other species, fNBCe1 is an electrogenic Na(+)/HCO(3)(-) cotransporter and sensitive to the stilbene inhibitor DIDS. However, our experiments revealed several unique and distinguishable fNBCe1 transport characteristics not found in mammalian or other teleost NBCe1-orthologs: electrogenic Li(+)/nHCO(3)(-) cotransport; HCO(3)(-) independent, DIDS-insensitive transport; and increased basal intracellular Na(+) accumulation. fNBCe1 is a voltage-dependent Na(+)/nHCO(3)(-) cotransporter that rectifies, independently from the extracellular Na(+) or HCO(3)(-) concentration, around -60 mV. Na(+) removal (0Na(+) prepulse) is necessary to produce the true HCO(3)(-)-elicited current. HCO(3)(-) addition results in huge outward currents with quick current decay. Kinetic analysis of HCO(3)(-) currents reveals that fNBCe1 has a much higher transport capacity (higher maximum current) and lower affinity (higher K(m)) than human kidney NBCe1 (hkNBCe1) does in the physiological range (membrane potential = -80 mV; [HCO(3)(-)] = 10 mM). In this state, fNBCe1 is in favor of operating as transepithelial HCO(3)(-) secretion, opposite of hkNBCe1, from blood to the luminal side. Thus, fugu-NBCe1 represents the first ortholog-based tool to study amino acid substitutions in NBCe1 and how those change ion and voltage dependence.


Asunto(s)
Simportadores de Sodio-Bicarbonato/metabolismo , Takifugu/fisiología , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Sustitución de Aminoácidos/efectos de los fármacos , Animales , Bicarbonatos/metabolismo , Transporte Biológico/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Femenino , Humanos , Cinética , Litio/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Sodio/metabolismo , Simportadores de Sodio-Bicarbonato/genética , Takifugu/genética , Takifugu/metabolismo , Xenopus laevis
10.
J Biol Chem ; 284(41): 28306-28318, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19643730

RESUMEN

SLC26 proteins function as anion exchangers, channels, and sensors. Previous cellular studies have shown that Slc26a3 and Slc26a6 interact with the R-region of the cystic fibrosis transmembrane conductance regulator (CFTR), (R)CFTR, via the Slc26-STAS (sulfate transporter anti-sigma) domain, resulting in mutual transport activation. We recently showed that Slc26a9 has both nCl(-)-HCO(3)(-) exchanger and Cl(-) channel function. In this study, we show that the purified STAS domain of Slc26a9 (a9STAS) binds purified (R)CFTR. When Slc26a9 and (R)CFTR fragments are co-expressed in Xenopus oocytes, both Slc26a9-mediated nCl(-)-HCO(3)(-) exchange and Cl(-) currents are almost fully inhibited. Deletion of the Slc26a9 STAS domain (a9-DeltaSTAS) virtually eliminated the Cl(-) currents with only a modest affect on nCl(-)-HCO(3)(-) exchange activity. Co-expression of a9-DeltaSTAS and the (R)CFTR fragment did not alter the residual a9-DeltaSTAS function. Replacing the Slc26a9 STAS domain with the Slc26a6 STAS domain (a6-a9-a6) does not change Slc26a9 function and is no longer inhibited by (R)CFTR. These data indicate that the Slc26a9-STAS domain, like other Slc26-STAS domains, binds CFTR in the R-region. However, unlike previously reported data, this binding interaction inhibits Slc26a9 ion transport activity. These results imply that Slc26-STAS domains may all interact with (R)CFTR but that the physiological outcome is specific to differing Slc26 proteins, allowing for dynamic and acute fine tuning of ion transport for various epithelia.


Asunto(s)
Antiportadores/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Animales , Antiportadores/genética , Bicarbonatos/metabolismo , Cloruros/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/metabolismo , Femenino , Humanos , Técnicas de Placa-Clamp , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transportadores de Sulfato , Xenopus laevis
11.
Am J Physiol Cell Physiol ; 297(4): C865-75, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19625604

RESUMEN

Mutations in the electrogenic Na+/nHCO3- cotransporter (NBCe1, SLC4A4) cause severe proximal renal tubular acidosis, glaucoma, and cataracts in humans, indicating NBCe1 has a critical role in acid-base homeostasis and ocular fluid transport. To better understand the homeostatic roles and protein ontogeny of NBCe1, we have cloned, localized, and downregulated NBCe1 expression in zebrafish, and examined its transport characteristics when expressed in Xenopus oocytes. Zebrafish NBCe1 (zNBCe1) is 80% identical to published mammalian NBCe1 cDNAs. Like other fish NBCe1 clones, zebrafish NBCe1 is most similar to the pancreatic form of mammalian NBC (Slc4a4-B) but appears to be the dominant isoform found in zebrafish. In situ hybridization of embryos demonstrated mRNA expression in kidney pronephros and eye by 24 h postfertilization (hpf) and gill and brain by 120 hpf. Immunohistochemical labeling demonstrated expression in adult zebrafish eye and gill. Morpholino knockdown studies demonstrated roles in eye and brain development and caused edema, indicating altered fluid and electrolyte balance. With the use of microelectrodes to measure membrane potential (Vm), voltage clamp (VC), intracellular pH (pH(i)), or intracellular Na+ activity (aNa(i)), we examined the function of zNBCe1 expressed in Xenopus oocytes. Zebrafish NBCe1 shared transport properties with mammalian NBCe1s, demonstrating electrogenic Na+ and HCO3- transport as well as similar drug sensitivity, including inhibition by 4,4'-diiso-thiocyano-2,2'-disulfonic acid stilbene and tenidap. These data indicate that NBCe1 in zebrafish shares many characteristics with mammalian NBCe1, including tissue distribution, importance in systemic water and electrolyte balance, and electrogenic transport of Na+ and HCO3-. Thus zebrafish promise to be useful model system for studies of NBCe1 physiology.


Asunto(s)
Simportadores de Sodio-Bicarbonato/fisiología , Proteínas de Pez Cebra/fisiología , Secuencia de Aminoácidos , Animales , Clonación Molecular , Embrión no Mamífero/metabolismo , Femenino , Activación del Canal Iónico , Transporte Iónico , Datos de Secuencia Molecular , Mutación , Oocitos/metabolismo , Especificidad de Órganos , Técnicas de Placa-Clamp , Simportadores de Sodio-Bicarbonato/genética , Xenopus , Pez Cebra , Proteínas de Pez Cebra/genética
12.
J Membr Biol ; 228(3): 125-40, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19365592

RESUMEN

The SLC26 gene family encodes anion transporters with diverse functional attributes: (a) anion exchanger, (b) anion sensor, and (c) anion conductance (likely channel). We have cloned and studied Slc26a9, a paralogue expressed mostly in lung and stomach. Immunohistochemistry shows that Slc26a9 is present at apical and intracellular membranes of lung and stomach epithelia. Using expression in Xenopus laevis oocytes and ion-sensitive microelectrodes, we discovered that Slc26a9 has a novel function not found in any other Slc26 proteins: cation coupling. Intracellular pH and voltage measurements show that Slc26a9 is a nCl(-)-HCO(3)(-) exchanger, suggesting roles in gastric HCl secretion or pulmonary HCO(3)(-) secretion; Na(+) electrodes and uptakes reveal that Slc26a9 has a cation dependence. Single-channel measurements indicate that Slc26a9 displays discrete open and closed states. These experiments show that Slc26a9 has three discrete physiological modes: nCl(-)-HCO(3)(-) exchanger, Cl(-) channel, and Na(+)-anion cotransporter. Thus, the Slc26a9 transporter channel is uniquely suited for dynamic and tissue-specific physiology or regulation in epithelial tissues.


Asunto(s)
Proteínas de Transporte de Anión/fisiología , Antiportadores/genética , Antiportadores de Cloruro-Bicarbonato/genética , Canales de Sodio/fisiología , Animales , Antiportadores/biosíntesis , Antiportadores/inmunología , Bicarbonatos/metabolismo , Clonación Molecular , Femenino , Humanos , Ratones , Oocitos , Técnicas de Placa-Clamp , Transportadores de Sulfato , Distribución Tisular , Xenopus laevis
13.
Int Urol Nephrol ; 51(12): 2209-2226, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31576489

RESUMEN

Chronic kidney disease (CKD) is a worldwide health problem, because it is one of the most common complications of metabolic diseases including obesity and type 2 diabetes. Patients with CKD also develop other comorbidities, such as hypertension, hyperlipidemias, liver and cardiovascular diseases, gastrointestinal problems, and cognitive deterioration, which worsens their health. Therapy includes reducing comorbidities or using replacement therapy, such as peritoneal dialysis, hemodialysis, and organ transplant. Health care systems are searching for alternative treatments for CKD patients to mitigate or retard their progression. One new topic is the study of uremic toxins (UT), which are excessively produced during CKD as products of food metabolism or as a result of the loss of renal function that have a negative impact on the kidneys and other organs. High urea concentrations significantly modify the microbiota in the gut also, cause a decrease in bacterial strains that produce anti-inflammatory and fuel molecules and an increase in bacterial strains that can metabolize urea, but also produce UT, including indoxyl sulfate and p-cresol sulfate. UT activates several cellular processes that induce oxidative environments, inflammation, proliferation, fibrosis development, and apoptosis; these processes mainly occur in the gut, heart, and kidney. The study of the microbiota during CKD allowed for the implementation of therapy schemes to try to reduce the circulating concentrations of UT and reduce the damage. The objective of this review is to show an overview to know the main UT produced in end-stage renal disease patients, and how prebiotics and probiotics intervention acts as a helpful tool in CKD treatment.


Asunto(s)
Microbioma Gastrointestinal , Insuficiencia Renal Crónica/microbiología , Microbioma Gastrointestinal/fisiología , Humanos , Prebióticos , Probióticos , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/terapia , Toxinas Biológicas/biosíntesis , Uremia/complicaciones , Uremia/metabolismo
14.
Novartis Found Symp ; 273: 126-38; discussion 138-47, 261-4, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17120765

RESUMEN

SLC26 anion exchangers transport monovalent and divalent anions, with a diversity of anion specificity and stoichiometry. Our microelectrode studies indicate that several SLC26 members are electrogenic. We reported that Slc26a6 functions as a Cl-/formate, Cl-/oxalate, Cl-/OH- and electrogenic Cl-/nHCO3- exchanger. Recently, we have also confirmed that Slc26a7 does not behave as a Cl-/HCO3- exchanger but does function as an electrogenic anion conductance, perhaps a channel. We have also cloned murine Slc26a9, which is strongly expressed in the respiratory tract and stomach. Radioisotope uptakes in Xenopus oocytes indicate that Slc26a9 is a highly selective anion exchanger, transporting Cl- but neither formate, oxalate, nor SO42-. We also utilized electrophysiology to voltage clamp (VC) and/or measure intracellular pH (pHi), Cl- ([Cl-],) and Na+ ([Na+]i), in response to various ion replacements. Cl- removal in HCO3- depolarizes oocytes (to > +60mV), alkalinizes oocytes, and decreases aCl-i. Slc26a9 thus functions as an electrogenic nCl-/HCO3- exchanger, suggesting a role in pulmonary and gastric HCO3- secretion and/or CO2 transport. VC experiments revealed channel-like currents (>10 microA at -60mV and >80 microA at +60mV) mediated by Slc26a9 in the presence and absence of HCO3-. Our experiments and those of others continue to reveal additional characteristics and unique roles for this new class of electrogenic anion transporters.


Asunto(s)
Antiportadores/metabolismo , Homología de Secuencia , Animales , Antiportadores/genética , Cloruros/metabolismo , Clonación Molecular , Transporte Iónico , Ratones , Modelos Biológicos , Oocitos , Técnicas de Placa-Clamp , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estómago/citología , Transportadores de Sulfato
15.
J Biol Chem ; 282(16): 11996-2009, 2007 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-17255103

RESUMEN

We have identified and characterized two different sodium-coupled monocarboxylate cotransporters (SMCT) from zebrafish (Danio rerio), electrogenic (zSMCTe) and electroneutral (zSMCTn). zSMCTn is the 12th member of the zebrafish Slc5 gene family (zSlc5a12). Both zSMCT sequences have approximately 50% homology to human SLC5A8 (hSMCT). Transport function and kinetics were measured in Xenopus oocytes injected with zSMCT cRNAs by measurement of intracellular Na(+) concentration ([Na(+)](i)) and membrane potential. Both zSMCTs oocytes increased [Na(+)](i) with addition of monocarboxylates (MC) such as lactate, pyruvate, nicotinate, and butyrate. By using two electrode voltage clamp experiments, we measured currents elicited from zSMCTe after MC addition. MC-elicited currents from zSMCTe were similar to hSMCT currents. In contrast, we found no significant MC-elicited current in either zSMCTn or control oocytes. Kinetic data show that zSMCTe has a higher affinity for lactate, nicotinate, and pyruvate (K(m)(L-lactate) = 0.17 +/- 0.02 mM, K(m)(nicotinate) = 0.54 +/- 0.12 mM at -150 mV) than zSMCTn (K(m)(L-lactate) = 1.81 +/- 0.19 mM, K(m)(nicotinate) = 23.68 +/- 4.88 mM). In situ hybridization showed that 1-, 3-, and 5-day-old zebrafish embryos abundantly express both zSMCTs in the brain, eyes, intestine, and kidney. Within the kidney, zSMCTn mRNA is expressed in pronephric tubules, whereas zSMCTe mRNA is more distal in pronephric ducts. zSMCTn is expressed in exocrine pancreas, but zSMCTe is not. Roles for Na(+)-coupled monocarboxylate cotransporters have not been described for the brain or eye. In summary, zSMCTe is the zebrafish SLC5A8 ortholog, and zSMCTn is a novel, electroneutral SMCT (zSlc5a12). Slc5a12 in higher vertebrates is likely responsible for the electroneutral Na(+)/lactate cotransport reported in mammalian and amphibian kidneys.


Asunto(s)
Proteínas de Transporte de Catión/fisiología , Transportadores de Ácidos Monocarboxílicos/fisiología , Sodio/metabolismo , Proteínas de Pez Cebra/fisiología , Animales , Electrofisiología , Humanos , Riñón/metabolismo , Cinética , Modelos Biológicos , Datos de Secuencia Molecular , Transportadores de Ácidos Monocarboxílicos/genética , Neuronas/metabolismo , Oocitos/metabolismo , Simportadores , Distribución Tisular , Xenopus , Pez Cebra , Proteínas de Pez Cebra/genética
16.
Am J Physiol Renal Physiol ; 290(5): F1094-102, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16291577

RESUMEN

The renal-specific Na(+)-K(+)-2Cl(-) cotransporter NKCC2 belongs to the SLC12 gene family; it is the target for loop diuretics and the cause of type I Bartter's syndrome. Because the NKCC2 sequence contains two putative N-linked glycosylation sites, one of which is conserved with the renal Na(+)-Cl(-) cotransporter in which glycosylation affects thiazide affinity, we assessed the role of glycosylation on NKCC2 functional properties. One (N442Q or N452Q) or both (N442,452Q) N-glycosylation sites were eliminated by site-directed mutagenesis. Wild-type NKCC2 and mutant clones were expressed in Xenopus laevis oocytes and analyzed by (86)Rb(+) influx, Western blotting, and confocal microscopy. Inhibition of glycosylation with tunicamycin in wild-type NKCC2-injected oocytes resulted in an 80% reduction of NKCC2 activity. Immunoblot of injected oocytes revealed that glycosylation of NKCC2 was completely prevented in N442,452Q-injected oocytes. Functional activity was reduced by 50% in N442Q- and N452Q-injected oocytes and by 80% in oocytes injected with N442,452Q, whereas confocal microscopy of oocytes injected with wild-type or mutant enhanced green fluorescent protein-tagged NKCC2 clones revealed that surface fluorescence intensity was reduced approximately 20% in single mutants and 50% in the double mutant. Ion transport kinetic analyses revealed no changes in cation affinity and a small increase in Cl(-) affinity by N442Q and N442,452Q. However, a slight decrease in bumetanide affinity was observed. Our data demonstrate that NKCC2 is glycosylated and suggest that prevention of glycosylation reduces its functional expression by affecting insertion into the plasma membrane and the intrinsic activity of the cotransporter.


Asunto(s)
Canales de Cloruro/fisiología , Asa de la Nefrona/fisiología , Simportadores de Cloruro de Sodio-Potasio/fisiología , Animales , Western Blotting , Membrana Celular/fisiología , Clonación Molecular , Electrofisiología , Glicosilación , Immunoblotting , Cinética , Microscopía Confocal , Mutagénesis Sitio-Dirigida , Oocitos , Isoformas de Proteínas , Ratas , Simportadores de Cloruro de Sodio-Potasio/biosíntesis , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 1 de la Familia de Transportadores de Soluto 12 , Xenopus laevis
17.
Am J Physiol Regul Integr Comp Physiol ; 289(5): R1520-34, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15994375

RESUMEN

In mammals, the Na+/H+ exchanger 3 (NHE3) is expressed with Na+/K+-ATPase in renal proximal tubules, where it secretes H+ and absorbs Na+ to maintain blood pH and volume. In elasmobranchs (sharks, skates, and stingrays), the gills are the dominant site of pH and osmoregulation. This study was conducted to determine whether epithelial NHE homologs exist in elasmobranchs and, if so, to localize their expression in gills and determine whether their expression is altered by environmental salinity or hypercapnia. Degenerate primers and RT-PCR were used to deduce partial sequences of mammalian NHE2 and NHE3 homologs from the gills of the euryhaline Atlantic stingray (Dasyatis sabina). Real-time PCR was then used to demonstrate that mRNA expression of the NHE3 homolog increased when stingrays were transferred to low salinities but not during hypercapnia. Expression of the NHE2 homolog did not change with either treatment. Rapid amplification of cDNA was then used to deduce the complete sequence of a putative NHE3. The 2,744-base pair cDNA includes a coding region for a 2,511-amino acid protein that is 70% identical to human NHE3 (SLC9A3). Antisera generated against the carboxyl tail of the putative stingray NHE3 labeled the apical membranes of Na+/K+-ATPase-rich epithelial cells, and acclimation to freshwater caused a redistribution of labeling in the gills. This study provides the first NHE3 cloned from an elasmobranch and is the first to demonstrate an increase in gill NHE3 expression during acclimation to low salinities, suggesting that NHE3 can absorb Na+ from ion-poor environments.


Asunto(s)
Branquias/metabolismo , Rajidae/metabolismo , Intercambiadores de Sodio-Hidrógeno/química , Intercambiadores de Sodio-Hidrógeno/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Branquias/citología , Inmunohistoquímica , Hibridación in Situ , Modelos Biológicos , Datos de Secuencia Molecular , Filogenia , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido
18.
J Biol Chem ; 277(13): 11004-12, 2002 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-11790783

RESUMEN

The bumetanide-sensitive Na(+):K(+):2Cl(-) cotransporter (BSC1) is the major pathway for salt reabsorption in the apical membrane of the mammalian thick ascending limb of Henle. Three isoforms of the cotransporter, known as A, B, and F, exhibit axial expression along the thick ascending limb. We report here a functional comparison of the three isoforms from mouse kidney. When expressed in Xenopus oocytes the mBSC1-A isoform showed higher capacity of transport, with no difference in the amount of surface expression. Kinetic characterization revealed divergent affinities for the three cotransported ions. The observed EC(50) values for Na(+), K(+), and Cl(-) were 5.0 +/- 3.9, 0.96 +/- 0.16, and 22.2 +/- 4.8 mm for mBSC1-A; 3.0 +/- 0.6, 0.76 +/- 0.07, and 11.6 +/- 0.7 mm for mBSC1-B; and 20.6 +/- 7.2, 1.54 +/- 0.16, and 29.2 +/- 2.1 mm for mBSC1-F, respectively. Bumetanide sensitivity was higher in mBSC1-B compared with the mBSC1-A and mBSC1-F isoforms. All three transporters were partially inhibited by hypotonicity but to different extents. The cell swelling-induced inhibition profile was mBSC1-F > mBSC1-B > mBSC1-A. The function of the Na(+):K(+):2Cl(-) cotransporter was not affected by extracellular pH or by the addition of metolazone, 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS), or R(+)-[(2-n-butyl-6,7-dichloro-2-cyclopentyl-2,3-dihydro-1-oxo-1-H-indenyl-5-yl)-oxy]acetic acid (DIOA) to the extracellular medium. In contrast, exposure of oocytes to HgCl(2) before the uptake period reduced the activity of the cotransporter. The effect of HgCl(2) was dose-dependent, and mBSC1-A and mBSC1-B exhibited higher affinity than mBSC1-F. Overall, the functional comparison of the murine apical renal-specific Na(+):K(+):2Cl(-) cotransporter isoforms A, B, and F reveals important functional, pharmacological, and kinetic differences, with both physiological and structural implications.


Asunto(s)
Isoformas de Proteínas/fisiología , Simportadores de Cloruro de Sodio-Potasio/fisiología , Secuencia de Aminoácidos , Animales , Bumetanida/farmacología , Femenino , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Concentración Osmolar , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico , Simportadores de Cloruro de Sodio-Potasio/química , Xenopus laevis
19.
Am J Physiol Renal Physiol ; 284(6): F1145-54, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12604467

RESUMEN

The murine apical bumetanide-sensitive Na(+)-K(+)-2Cl(-) cotransporter gene (mBSC1) exhibits two spliced isoform products that differ at the COOH-terminal domain. A long COOH-terminal isoform (L-mBSC1) encodes the Na(+)-K(+)-2Cl(-) cotransporter, and a short isoform (S-mBSC1) exerts a dominant-negative effect on L-mBSC1 cotransporter activity that is abrogated by cAMP. However, the mechanism of this dominant-negative effect was not clear. In this study, we used confocal microscopic analysis of an enhanced green fluorescent protein (EGFP) fusion construct (L-mBSC1-EGFP) expressed to characterize the surface expression of the L-BSC1 isoform in Xenopus laevis oocytes. Functional expression was also assessed in L-mBSC1-injected oocytes by measuring the bumetanide-sensitive (86)Rb(+) uptake. Oocytes injected with L-mBSC1-EGFP cRNA developed a distinct plasma membrane-associated fluorescence that colocalized with the fluorescent membrane dye FM 4-64. The fluorescence intensity in L-mBSC1-EGFP oocytes did not change after cAMP was added to the extracellular medium. In contrast, L-mBSC1-EGFP fluorescence intensity was reduced in a dose-dependent manner, with coexpression of S-mBSC1. The inhibitory effect of S-mBSC1 was abrogated by cAMP. Finally, the exocytosis inhibitor colchicine blocked the effect of cAMP on the L-mBSC1-EGFP/S-mBSC1-coinjected oocytes. All changes in L-mBSC1 surface expression correlated with modification of bumetanide-sensitive (86)Rb(+) uptake. Our data suggest that the dominant-negative effect of S-mBSC1 on L-mBSC1 transport function is due to the effects of the cotransporter on trafficking.


Asunto(s)
AMP Cíclico/fisiología , Riñón/enzimología , Riñón/fisiología , Simportadores de Cloruro de Sodio-Potasio/metabolismo , 1-Metil-3-Isobutilxantina/farmacología , Animales , Bucladesina/antagonistas & inhibidores , Bucladesina/farmacología , Membrana Celular/enzimología , Membrana Celular/metabolismo , Colchicina/farmacología , ADN Complementario/biosíntesis , Activación Enzimática , Exocitosis/fisiología , Proteínas Fluorescentes Verdes , Técnicas In Vitro , Isoenzimas/biosíntesis , Isoenzimas/metabolismo , Proteínas Luminiscentes , Proteínas de la Membrana/biosíntesis , Ratones , Oocitos , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación , Simportadores de Cloruro de Sodio-Potasio/biosíntesis , Xenopus
20.
Am J Physiol Renal Physiol ; 287(3): F570-7, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15149970

RESUMEN

The mammalian kidney bumetanide-sensitive Na(+)-K(+)-2Cl(-) and thiazide-sensitive Na(+)-Cl(-) cotransporters are the major pathways for salt reabsorption in the thick ascending limb of Henle's loop and distal convoluted tubule, respectively. These cotransporters serve as receptors for the loop- and thiazide-type diuretics, and inactivating mutations of corresponding genes are associated with development of Bartter's syndrome type I and Gitleman's disease, respectively. Structural requirements for ion translocation and diuretic binding specificity are unknown. As an initial approach for analyzing structural determinants conferring ion or diuretic preferences in these cotransporters, we exploited functional differences and structural similarities between Na(+)-K(+)-2Cl(-) and Na(+)-Cl(-) cotransporters to design and study chimeric proteins in which the NH(2)-terminal and/or COOH-terminal domains were switched between each other. Thus six chimeric proteins were produced. Using the heterologous expression system of Xenopus laevis oocytes, we observed that four chimeras exhibited functional activity. Our results revealed that, in the Na(+)-K(+)-2Cl(-) cotransporter, ion translocation and diuretic binding specificity are determined by the central hydrophobic domain. Thus NH(2)-terminal and COOH-terminal domains do not play a role in defining these properties. A similar conclusion can be suggested for the Na(+)-Cl(-) cotransporter.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Asa de la Nefrona/metabolismo , Receptores de Droga/genética , Receptores de Droga/metabolismo , Simportadores de Cloruro de Sodio-Potasio/genética , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Simportadores , Animales , Benzotiadiazinas , Bumetanida/farmacología , Proteínas Portadoras/química , Polaridad Celular/fisiología , Clonación Molecular , Diuréticos/farmacología , Femenino , Mutagénesis , Oocitos , Estructura Terciaria de Proteína , Ratas , Receptores de Droga/química , Proteínas Recombinantes de Fusión/metabolismo , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Simportadores del Cloruro de Sodio , Simportadores de Cloruro de Sodio-Potasio/química , Miembro 1 de la Familia de Transportadores de Soluto 12 , Miembro 3 de la Familia de Transportadores de Soluto 12 , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA