Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Silvana Bazúa-Valenti is active.

Publication


Featured researches published by Silvana Bazúa-Valenti.


Journal of The American Society of Nephrology | 2015

The Effect of WNK4 on the Na+–Cl− Cotransporter Is Modulated by Intracellular Chloride

Silvana Bazúa-Valenti; María Chávez-Canales; Lorena Rojas-Vega; Xochiquetzal Gonzalez-Rodriguez; Norma Vázquez; Alejandro Rodríguez-Gama; Eduardo R. Argaiz; Zesergio Melo; Consuelo Plata; David H. Ellison; Jesus Garcia-Valdes; Juliette Hadchouel; Gerardo Gamba

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.


American Journal of Physiology-cell Physiology | 2015

Revisiting the NaCl cotransporter regulation by with-no-lysine kinases

Silvana Bazúa-Valenti; Gerardo Gamba

The renal thiazide-sensitive Na(+)-Cl(-) cotransporter (NCC) is the salt transporter in the distal convoluted tubule. Its activity is fundamental for defining blood pressure levels. Decreased NCC activity is associated with salt-remediable arterial hypotension with hypokalemia (Gitelman disease), while increased activity results in salt-sensitive arterial hypertension with hyperkalemia (pseudohypoaldosteronism type II; PHAII). The discovery of four different genes causing PHAII revealed a complex multiprotein system that regulates the activity of NCC. Two genes encode for with-no-lysine (K) kinases WNK1 and WNK4, while two encode for kelch-like 3 (KLHL3) and cullin 3 (CUL3) proteins that form a RING type E3 ubiquitin ligase complex. Extensive research has shown that WNK1 and WNK4 are the targets for the KLHL3-CUL3 complex and that WNKs modulate the activity of NCC by means of intermediary Ste20-type kinases known as SPAK or OSR1. The understanding of the effect of WNKs on NCC is a complex issue, but recent evidence discussed in this review suggests that we could be reaching the end of the dark ages regarding this matter.


American Journal of Kidney Diseases | 2017

The Effect of Spironolactone on Acute Kidney Injury After Cardiac Surgery: A Randomized, Placebo-Controlled Trial

Rubén Barba-Navarro; Mirell Tapia-Silva; Carlos Garza-Garcia; Salvador López-Giacoman; Ipsae Melgoza-Toral; Armando Vazquez-Rangel; Silvana Bazúa-Valenti; Norma A. Bobadilla; Michael Wasung de Lay; Francisco Baranda; Lakhmir S. Chawla; Gerardo Gamba; Magdalena Madero

BACKGROUND Cardiac surgery-related acute kidney injury (AKI) is a common postoperative complication that greatly increases morbidity and mortality. There are currently no effective interventions to prevent AKI associated with cardiac surgery. Experimental data have shown that administration of the mineralocorticoid receptor blocker spironolactone prevents renal injury induced by ischemia-reperfusion in rats. The objective of this study was to test whether short-term perioperative administration of oral spironolactone could reduce the incidence of AKI in cardiac surgical patients. STUDY DESIGN Randomized, double-blinded, placebo-controlled trial. SETTING & PARTICIPANTS Data were collected from April 2014 through July 2015 at the National Heart Institute in Mexico. 233 patients were included; 115 and 118 received spironolactone or placebo, respectively. INTERVENTION Spironolactone or placebo once at a dose of 100mg 12 to 24 hours before surgery and subsequently 3 further doses of 25mg in postoperative days 0, 1, and 2 were administered. OUTCOMES Patients were followed up for 7 days or until discharge from the intensive care unit (ICU). The primary end point was AKI incidence defined by KDIGO criteria. Secondary end points included requirement of renal replacement therapy, ICU length of stay, and ICU mortality. Data were analyzed according to the intention-to-treat principle. RESULTS Mean age was 53.2±15 years, mean serum creatinine level was 0.9±0.2mg/dL, median Thakar score for estimation of AKI risk was 2 (IQR, 1-3), and 25% had diabetes. The incidence of AKI was higher for the spironolactone group (43% vs 29%; P=0.02). No significant differences were found for secondary end points. LIMITATIONS Single center, AKI was mostly driven by AKI stage 1, planned sample size was not achieved, and there was no renin-angiotensin-aldosterone system washout period. CONCLUSIONS Our trial demonstrated that spironolactone was not protective for AKI associated with cardiac surgery and there may be a trend toward risk.


Biochimica et Biophysica Acta | 2014

Isoforms of protein kinase C involved in phorbol ester-induced sphingosine 1-phosphate receptor 1 phosphorylation and desensitization.

Marco Antonio Morquecho-León; Silvana Bazúa-Valenti; M. Teresa Romero-Ávila; J. Adolfo García-Sáinz

The role of protein kinase C (PKC) isozymes in phorbol myristate acetate (PMA)-induced sphingosine 1-phosphate (S1P) receptor 1 (S1P1) phosphorylation was studied. Activation of S1P1 receptors induced an immediate increase in intracellular calcium, which was blocked by preincubation with PMA. Both S1P and PMA were able to increase S1P1 phosphorylation in a concentration- and time-dependent fashion. Down-regulation of PKC (overnight incubation with PMA) blocked the subsequent effect of the phorbol ester on S1P1 phosphorylation, without decreasing that of the natural agonist. Pharmacological inhibition of PKC α prevented the effects of PMA on S1P-triggered intracellular calcium increase and on S1P1 phosphorylation; no such effect was observed on the effects of the sphingolipid agonist. The presence of PKC α and β isoforms in S1P1 immunoprecipitates was evidenced by Western blotting. Additionally, expression of dominant-negative mutants of PKC α or β and knockdown of these isozymes using short hairpin RNA, markedly attenuated PMA-induced S1P1 phosphorylation. Our results indicate that the classical isoforms, mainly PKC α, mediate PMA-induced phosphorylation and desensitization of S1P1.


American Journal of Physiology-renal Physiology | 2015

Increased phosphorylation of the renal Na+-Cl− cotransporter in male kidney transplant recipient patients with hypertension: a prospective cohort

Lorena Rojas-Vega; Aldo R. Jiménez-Vega; Silvana Bazúa-Valenti; Isidora Arroyo-Garza; José Víctor Jiménez; Ruy Gómez-Ocádiz; Diego Luis Carrillo-Pérez; Erika Moreno; Luis E. Morales-Buenrostro; Josefina Alberú; Gerardo Gamba

Evidence in rodents suggests that tacrolimus-induced posttransplant hypertension is due to upregulation of the thiazide-sensitive Na+-Cl- cotransporter NCC. Here, we analyzed whether a similar mechanism is involved in posttransplant hypertension in humans. From January 2013 to June 2014, all adult kidney transplant recipients receiving a kidney allograft were enrolled in a prospective cohort study. All patients received tacrolimus as part of the immunosuppressive therapy. Six months after surgery, we assessed general clinical and laboratory variables, tacrolimus trough blood levels, and ambulatory 24-h blood pressure monitoring. Urinary exosomes were extracted to perform Western blot analysis using total and phospho-NCC antibodies. A total of 52 patients, including 17 women and 35 men, were followed. At 6 mo after transplantation, of the 35 men, 17 developed hypertension and 18 remained normotensive, while high blood pressure was observed in only 3 of 17 women. The hypertensive patients were significantly older than the normotensive group; however, there were no significant differences in body weight, history of acute rejection, renal function, and tacrolimus trough levels. In urinary exosomes, hypertensive patients showed higher NCC expression (1.7±0.19) than normotensive (1±0.13) (P=0.0096). Also, NCC phosphorylation levels were significantly higher in the hypertensive patients (1.57±0.16 vs. 1±0.07; P=0.0049). Our data show that there is a positive correlation between NCC expression/phosphorylation in urinary exosomes and the development of hypertension in posttransplant male patients treated with tacrolimus. Our results are consistent with the hypothesis that NCC activation plays a major role in tacrolimus-induced hypertension.


American Journal of Physiology-renal Physiology | 2016

Physiological role of SLC12 family members in the kidney

Silvana Bazúa-Valenti; María Castañeda-Bueno; Gerardo Gamba

The solute carrier family 12, as numbered according to Human Genome Organisation (HUGO) nomenclature, encodes the electroneutral cation-coupled chloride cotransporters that are expressed in many cells and tissues; they play key roles in important physiological events, such as cell volume regulation, modulation of the intracellular chloride concentration, and transepithelial ion transport. Most of these family members are expressed in specific regions of the nephron. The Na-K-2Cl cotransporter NKCC2, which is located in the thick ascending limb, and the Na-Cl cotransporter, which is located in the distal convoluted tubule, play important roles in salt reabsorption and serve as the receptors for loop and thiazide diuretics, respectively (Thiazide diuretics are among the most commonly prescribed drugs in the world.). The activity of these transporters correlates with blood pressure levels; thus, their regulation has been a subject of intense research for more than a decade. The K-Cl cotransporters KCC1, KCC3, and KCC4 are expressed in several nephron segments, and their role in renal physiology is less understood but nevertheless important. Evidence suggests that they are involved in modulating proximal tubule glucose reabsorption, thick ascending limb salt reabsorption and collecting duct proton secretion. In this work, we present an overview of the physiological roles of these transporters in the kidney, with particular emphasis on the knowledge gained in the past few years.


Journal of The American Society of Nephrology | 2018

The Calcium-Sensing Receptor Increases Activity of the Renal NCC through the WNK4-SPAK Pathway

Silvana Bazúa-Valenti; Lorena Rojas-Vega; María Castañeda-Bueno; Jonatan Barrera-Chimal; Rocio Bautista; Luz Graciela Cervantes-Pérez; Norma Vázquez; Consuelo Plata; Adrián Rafael Murillo-de-Ozores; Lorenza González-Mariscal; David H. Ellison; Daniela Riccardi; Norma A. Bobadilla; Gerardo Gamba

Background Hypercalciuria can result from activation of the basolateral calcium-sensing receptor (CaSR), which in the thick ascending limb of Henles 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.


Journal of Biological Chemistry | 2018

C-terminally truncated, kidney-specific variants of the WNK4 kinase lack several sites that regulate its activity

Adrián Rafael Murillo-de-Ozores; Alejandro Rodríguez-Gama; Silvana Bazúa-Valenti; Karla Leyva-Ríos; Norma Vázquez; Diana Pacheco-Alvarez; Inti A. De La Rosa-Velázquez; Agnieszka Wengi; Kathryn L. Stone; Junhui Zhang; Johannes Loffing; Richard P. Lifton; Chao Ling Yang; David H. Ellison; Gerardo Gamba; María Castañeda-Bueno

WNK lysine-deficient protein kinase 4 (WNK4) is an important regulator of renal salt handling. Mutations in its gene cause pseudohypoaldosteronism type II, mainly arising from overactivation of the renal Na+/Cl− cotransporter (NCC). In addition to full-length WNK4, we have observed faster migrating bands (between 95 and 130 kDa) in Western blots of kidney lysates. Therefore, we hypothesized that these could correspond to uncharacterized WNK4 variants. Here, using several WNK4 antibodies and WNK4−/− mice as controls, we showed that these bands indeed correspond to short WNK4 variants that are not observed in other tissue lysates. LC-MS/MS confirmed these bands as WNK4 variants that lack C-terminal segments. In HEK293 cells, truncation of WNK4s C terminus at several positions increased its kinase activity toward Ste20-related proline/alanine-rich kinase (SPAK), unless the truncated segment included the SPAK-binding site. Of note, this gain-of-function effect was due to the loss of a protein phosphatase 1 (PP1)-binding site in WNK4. Cotransfection with PP1 resulted in WNK4 dephosphorylation, an activity that was abrogated in the PP1-binding site WNK4 mutant. The electrophoretic mobility of the in vivo short variants of renal WNK4 suggested that they lack the SPAK-binding site and thus may not behave as constitutively active kinases toward SPAK. Finally, we show that at least one of the WNK4 short variants may be produced by proteolysis involving a Zn2+-dependent metalloprotease, as recombinant full-length WNK4 was cleaved when incubated with kidney lysate.


Archive | 2018

The calcium-sensing receptor increases activity of the renal NaCl cotransporter through the WNK4-SPAK pathway

Silvana Bazúa-Valenti; Lorena Rojas-Vega; María Castañeda-Bueno; Jonatan Barrera-Chimal; Rocio Bautista; Luz Graciela Cervantes-Pérez; Norma Vázquez; Consuelo Plata; Adrian R. Murillo-de-Ozpres; Lorenza González-Mariscal; David H. Ellison; Daniela Riccardi; Norma A. Bobadilla; Gerardo Gamba


The FASEB Journal | 2015

The Effect of WNK4 on the Na+:Cl- Cotransporter is modulated by Intracellular Chloride

Silvana Bazúa-Valenti; María Chávez-Canales; Lorena Rojas-Vega; Xochiquetzal Gonzalez-Rodriguez; Norma Vázquez; Alejandro Rodríguez-Gama; Eduardo R. Argaiz; Consuelo Plata; Jesus Garcia-Valdes; David H. Ellison; Juliette Hadchouel; Gerardo Gamba

Collaboration


Dive into the Silvana Bazúa-Valenti's collaboration.

Top Co-Authors

Avatar

Gerardo Gamba

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lorena Rojas-Vega

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Norma Vázquez

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Consuelo Plata

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

María Castañeda-Bueno

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Alejandro Rodríguez-Gama

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Norma A. Bobadilla

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Adrián Rafael Murillo-de-Ozores

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Eduardo R. Argaiz

National Autonomous University of Mexico

View shared research outputs
Researchain Logo
Decentralizing Knowledge