5 research outputs found

    Sprouty1 controls genitourinary development via its N-terminal tyrosine

    Get PDF
    Background: Congenital anomalies of the kidney and urinary tract (CAKUT) is a group of diseases that include a broad spectrum of developmental defects of the genitourinary system. Mouse models indicate that perturbations of the GDNF-Ret signaling pathway are a major genetic cause of CAKUT. Sprouty1 is an intracellular Ret inhibitor whose mutation results in supernumerary kidneys, megaureters, and hydronephrosis in mice. Both the molecular mechanisms and the structural domains critical for Sprouty function are a matter of controversy, partly because studies pursuing this objective rely on ectopic overexpression in cell lines. A conserved N-terminal tyrosine has been frequently, but not always, identified as critical for their function in vitro. Methods: We have generated Sprouty1 knockin mice bearing a tyrosine-to-alanine substitution in position 53, corresponding to the conserved N-terminal tyrosine of Sprouty1. We have characterized development of the genitourinary systems of these mice via different methods, including the use of reporter mice expressing EGFP form the Ret locus, and whole mount cytokeratin staining. Results: Mice lacking this tyrosine grow ectopic ureteric buds that ultimately will form supernumerary kidneys, a phenotype indistinguishable to that of Sprouty1 knockout mice. Sprouty1 knockin mice also present megaureters and vesicoureteral reflux, caused by failure of ureters to separate from Wolffian ducts and migrate to their definitive position. Conclusions: Tyrosine 53 is absolutely necessary to convey Sprouty1 function during genitourinary development.This work was supported by grants BFU2010-47175-P and BFU2017-83646-P (AEI/FEDER, UE) from MINECO to ME. MV was supported by a predoctoral fellowship from AGAUR. CA was supported by a predoctoral fellowship from Universitat de Lleida. SC was supported by a Cofund action from the Marie Curie program of the EU. We are grateful to Dr. Sanjay Jain (Washington University, St Louis) for sharing RetEGFP mice, and to Dr. Tung-Tien Sun (New York University) for Uroplakin antibody. We thank Anna Macià (IRB Lleida) for her contribution to the initial development of this manuscript, as well as Marta Hereu, Maria Santacana, Mónica Domingo and Maria Carrele for their excellent technical assistance

    A dominant negative mutation uncovers cooperative control of caudal Wolffian duct development by Sprouty genes

    Get PDF
    The Wolffian ducts (WD) are paired epithelial tubules central to the development of the mammalian genitourinary tract. Outgrowths from the WD known as the ureteric buds (UB) generate the collecting ducts of the kidney. Later during development, the caudal portion of the WD will form the vas deferens, epididymis and seminal vesicle in males, and will degenerate in females. While the genetic pathways controlling the development of the UB are firmly established, less is known about those governing development of WD portions caudal to the UB. Sprouty proteins are inhibitors of receptor tyrosine kinase (RTK) signaling in vivo. We have recently shown that homozygous mutation of a conserved tyrosine (Tyr53) of Spry1 results in UB defects indistinguishable from that of Spry1 null mice. Here, we show that heterozygosity for the Spry1 Y53A allele causes caudal WD developmental defects consisting of ectopically branched seminal vesicles in males and persistent WD in females, without affecting kidney development. Detailed analysis reveals that this phenotype also occurs in Spry1+/- mice but with a much lower penetrance, indicating that removal of tyrosine 53 generates a dominant negative mutation in vivo. Supporting this notion, concomitant deletion of one allele of Spry1 and Spry2 also recapitulates the genital phenotype of Spry1Y53A/+ mice with high penetrance. Mechanistically, we show that unlike the effects of Spry1 in kidney development, these caudal WD defects are independent of Ret signaling, but can be completely rescued by lowering the genetic dosage of Fgf10. In conclusion, mutation of tyrosine 53 of Spry1 generates a dominant negative allele that uncovers fine-tuning of caudal WD development by Sprouty genes.Open Access funding provided thanks to the CRUE-CSIC agreement with Springer Nature. This work was supported by grants BFU2017-83646-P (MINECO) and PID2020-114947 GB-I00 (MCIU) (both supported by funds from AEI/FEDER, UE) to ME. MV was supported by a predoctoral fellowship from AGAUR. GA and CA and GA are supported by a fellowship from Universitat de Lleida. SC was supported by a Cofund action from the Marie Curie program of the E

    Caracterización de la senescencia celular inducida por Sprouty: papel de p38

    Get PDF
    Els membres de la família de proteïnes Sprouty han estat clàssicament descrits com a inhibidors de la senyalització iniciada pels receptors tirosina quinasa, sent per això reiteradament proposats com supressors tumorals. D'altra banda, la senescència cel·lular és una resposta a certs nivells d'estrès en la qual les cèl·lules decideixen abandonar la seva capacitat proliferativa i secretar un ampli ventall de factors per influir sobre l'entorn proper. Treballs previs demostren que Spry1 promou la inducció de senescència cel·lular tant en tiroide com en el desenvolupament de la vagina de manera independent a la seva acció clàssica sobre els receptors tirosina quinasa. En la present tesi hem expandit la implicació de Spry1 en senescència a paradigmes en els quals aquesta és rellevant com l'envelliment, la reparació de teixits o la senescència associada a quimioteràpia. A més, hem revelat que Spry1 respon a l'estrès oxidatiu potenciant la via p38. Per tant, els resultats exposats confirmen a les proteïnes Spry com a mediadors generals de la senescència cel·lular.Los miembros de la familia de proteínas Sprouty han sido clásicamente descritos como inhibidores de la señalización iniciada por los receptores tirosina quinasa, siendo por ello reiteradamente propuestos como supresores tumorales. Por otro lado, la senescencia celular es una respuesta a ciertos niveles de estrés en la que las células deciden abandonar su capacidad proliferativa y secretar un amplio abanico de factores para influir sobre el entorno próximo. Trabajos previos demuestran que Spry1 promueve la inducción de senescencia celular tanto en tiroides como en el desarrollo de la vagina de manera independiente a su acción clásica sobre los receptores tirosina quinasa. En la presente tesis hemos expandido la implicación de Spry1 en senescencia a paradigmas en los que ésta es relevante como el envejecimiento, la reparación de tejidos o la senescencia asociada a quimioterapia. Además, hemos desvelado que Spry1 responde al estrés oxidativo potenciando la vía p38. Por tanto, los resultados expuestos confirman a las proteínas Spry como mediadores generales de la senescencia celular.Members of Sprouty family of genes (Spry1-4) have been classically assumed to be inhibitors of signaling initiated by receptor tyrosine kinases. As such, they have been proposed as tumor suppressor genes. On the other hand, cellular senescence is a response in front of different cellular stresses in which cells decide to cease their proliferation. Moreover, senescent cells implement the secretion of a myriad of factors to influence their proximal environment. Previous data from our group demonstrate that Spry1 promotes cellular senescence in the thyroid gland and in vaginal development, but in an ERK-independent manner. The present work expands the implications of Spry genes to other cellular senescence scenarios such as age-related and regeneration-related senescence. Surprisingly, Spry1 induction in the senescent response is controlled by oxidative stress. Mechanistically, Spry proteins enhance p38 MAPK activation required for the proper induction of most of the traits of cellular senescence. Hence, these results confirm Spry genes as novel, general mediators of cellular senescence

    Caracterización de la senescencia celular inducida por Sprouty: papel de p38

    No full text
    Els membres de la família de proteïnes Sprouty han estat clàssicament descrits com a inhibidors de la senyalització iniciada pels receptors tirosina quinasa, sent per això reiteradament proposats com supressors tumorals. D'altra banda, la senescència cel·lular és una resposta a certs nivells d'estrès en la qual les cèl·lules decideixen abandonar la seva capacitat proliferativa i secretar un ampli ventall de factors per influir sobre l'entorn proper. Treballs previs demostren que Spry1 promou la inducció de senescència cel·lular tant en tiroide com en el desenvolupament de la vagina de manera independent a la seva acció clàssica sobre els receptors tirosina quinasa. En la present tesi hem expandit la implicació de Spry1 en senescència a paradigmes en els quals aquesta és rellevant com l'envelliment, la reparació de teixits o la senescència associada a quimioteràpia. A més, hem revelat que Spry1 respon a l'estrès oxidatiu potenciant la via p38. Per tant, els resultats exposats confirmen a les proteïnes Spry com a mediadors generals de la senescència cel·lular.Los miembros de la familia de proteínas Sprouty han sido clásicamente descritos como inhibidores de la señalización iniciada por los receptores tirosina quinasa, siendo por ello reiteradamente propuestos como supresores tumorales. Por otro lado, la senescencia celular es una respuesta a ciertos niveles de estrés en la que las células deciden abandonar su capacidad proliferativa y secretar un amplio abanico de factores para influir sobre el entorno próximo. Trabajos previos demuestran que Spry1 promueve la inducción de senescencia celular tanto en tiroides como en el desarrollo de la vagina de manera independiente a su acción clásica sobre los receptores tirosina quinasa. En la presente tesis hemos expandido la implicación de Spry1 en senescencia a paradigmas en los que ésta es relevante como el envejecimiento, la reparación de tejidos o la senescencia asociada a quimioterapia. Además, hemos desvelado que Spry1 responde al estrés oxidativo potenciando la vía p38. Por tanto, los resultados expuestos confirman a las proteínas Spry como mediadores generales de la senescencia celular.Members of Sprouty family of genes (Spry1-4) have been classically assumed to be inhibitors of signaling initiated by receptor tyrosine kinases. As such, they have been proposed as tumor suppressor genes. On the other hand, cellular senescence is a response in front of different cellular stresses in which cells decide to cease their proliferation. Moreover, senescent cells implement the secretion of a myriad of factors to influence their proximal environment. Previous data from our group demonstrate that Spry1 promotes cellular senescence in the thyroid gland and in vaginal development, but in an ERK-independent manner. The present work expands the implications of Spry genes to other cellular senescence scenarios such as age-related and regeneration-related senescence. Surprisingly, Spry1 induction in the senescent response is controlled by oxidative stress. Mechanistically, Spry proteins enhance p38 MAPK activation required for the proper induction of most of the traits of cellular senescence. Hence, these results confirm Spry genes as novel, general mediators of cellular senescence
    corecore