Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Ther ; 28(8): 1818-1832, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32534604

RESUMEN

Asherman's syndrome (AS) is characterized by intrauterine adhesions or fibrosis resulting from scarring inside the endometrium. AS is associated with infertility, recurrent miscarriage, and placental abnormalities. Although mesenchymal stem cells show therapeutic promise for the treatment of AS, the molecular mechanisms underlying its pathophysiology remain unclear. We ascertained that mice with AS, like human patients with AS, suffer from extensive fibrosis, oligo/amenorrhea, and infertility. Human perivascular stem cells (hPVSCs) from umbilical cords repaired uterine damage in mice with AS, regardless of their delivery routes. In mice with AS, embryo implantation is aberrantly deferred, which leads to intrauterine growth restriction followed by no delivery at term. hPVSC administration significantly improved implantation defects and subsequent poor pregnancy outcomes via hypoxia inducible factor 1α (HIF1α)-dependent angiogenesis in a dose-dependent manner. Pharmacologic inhibition of HIF1α activity hindered hPVSC actions on pregnancy outcomes, whereas stabilization of HIF1α activity facilitated such actions. Furthermore, therapeutic effects of hPVSCs were not observed in uterine-specific HIF1α-knockout mice with AS. Secretome analyses of hPVSCs identified cyclophilin-A as the major paracrine factor for hPVSC therapy via HIF1α-dependent angiogenesis. Collectively, we demonstrate that hPVSCs-derived cyclophilin-A facilitates HIF1α-dependent angiogenesis to ameliorate compromised uterine environments in mice with AS, representing the major pathophysiologic features of humans with AS.


Asunto(s)
Ciclofilina A/biosíntesis , Ginatresia/etiología , Ginatresia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Madre Mesenquimatosas/metabolismo , Neovascularización Patológica/genética , Útero/metabolismo , Útero/patología , Animales , Biomarcadores , Biopsia , Modelos Animales de Enfermedad , Femenino , Fertilidad , Fibrosis , Ginatresia/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Comunicación Paracrina , Fenotipo , Regeneración
2.
Clin Exp Reprod Med ; 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38757275

RESUMEN

Objective: Autophagy is a major intracellular catabolic pathway governed by the sequential actions of proteins encoded by autophagy-related genes (Atg). ATG9, the only transmembrane protein involved in this process, regulates phospholipid translocation to autophagosomes during the early phases of autophagy. In mammals, two Atg9 isoforms have been reported: Atg9a and Atg9b. In this study, we examined whether the molecular and cellular characteristics of these two isoforms differed in mice. Methods: Whole uteri were collected on days 1, 4, and 8 of pregnancy and from ovariectomized mice injected with vehicle, progesterone, or 17ß-estradiol. Cells from reproductive tissues, such as granulosa cells, uterine epithelial cells (UECs), uterine stromal cells (USCs), and oocytes were collected. Two human uterine cell lines were also used in this analysis. Reverse transcription-polymerase chain reaction tests, Western blotting, and immunofluorescence staining were performed. Serum starvation conditions were used to induce autophagy in primary cells. Results: Atg9a and Atg9b were expressed in multiple mouse tissues and reproductive cells. Neither Atg9A nor Atg9B significantly changed in response to steroid hormones. Immunofluorescence staining of the UECs and USCs showed that ATG9A was distributed in a punctate-like pattern, whereas ATG9B exhibited a pattern of elongated tubular shapes in the cytoplasm. In human cancer cell lines, ATG9B was undetectable, whereas ATG9A was found in all cell types examined. Conclusion: The Atg9 isoforms exhibited distinct subcellular localizations in UECs and may play different roles in autophagy. Notably, human uterine cells exhibited reduced ATG9B expression, suggesting that this suppression may be due to epigenetic regulation.

3.
Nat Commun ; 14(1): 3220, 2023 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-37270588

RESUMEN

Progesterone (P4) is required for the preparation of the endometrium for a successful pregnancy. P4 resistance is a leading cause of the pathogenesis of endometrial disorders like endometriosis, often leading to infertility; however, the underlying epigenetic cause remains unclear. Here we demonstrate that CFP1, a regulator of H3K4me3, is required for maintaining epigenetic landscapes of P4-progesterone receptor (PGR) signaling networks in the mouse uterus. Cfp1f/f;Pgr-Cre (Cfp1d/d) mice showed impaired P4 responses, leading to complete failure of embryo implantation. mRNA and chromatin immunoprecipitation sequencing analyses showed that CFP1 regulates uterine mRNA profiles not only in H3K4me3-dependent but also in H3K4me3-independent manners. CFP1 directly regulates important P4 response genes, including Gata2, Sox17, and Ihh, which activate smoothened signaling pathway in the uterus. In a mouse model of endometriosis, Cfp1d/d ectopic lesions showed P4 resistance, which was rescued by a smoothened agonist. In human endometriosis, CFP1 was significantly downregulated, and expression levels between CFP1 and these P4 targets are positively related regardless of PGR levels. In brief, our study provides that CFP1 intervenes in the P4-epigenome-transcriptome networks for uterine receptivity for embryo implantation and the pathogenesis of endometriosis.


Asunto(s)
Endometriosis , Progesterona , Transactivadores , Animales , Femenino , Humanos , Ratones , Embarazo , Implantación del Embrión/genética , Endometriosis/genética , Endometriosis/metabolismo , Endometrio/metabolismo , Epigénesis Genética , Progesterona/farmacología , Progesterona/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , ARN Mensajero/metabolismo , Útero/metabolismo , Transactivadores/genética
4.
Mol Biol Rep ; 38(2): 833-40, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20376700

RESUMEN

The human COX-2 promoter contains a direct repeat 1 (DR1) which was shown to confer responsiveness to PPARs. We found that in AN(3)CA and F9 cells, this hCOX-2 DR1 mediates responsiveness to all-trans-retinoic acid (tRA) or 9-cis-retinoic acid (9cRA), but this effect was suppressed by PPARδ. Truncated PPARδ lacking the activation domain AF2 cannot suppress RA-induced activation of the hCOX-2 gene via DR1, suggesting that cofactor recruitment by AF2 is required for the suppression by PPARδ. Gel shift assay showed that PPAR/RXR, RARß/RXR, and RXR/RXR, bind to hCOX-2 DR1, revealing the promiscuity of this DR1. Particularly, RXR homodimer was able to bind to this DR1 only in the presence of 9cRA. Our results established that tRA and 9cRA are potent inducers of hCOX-2 and that the hCOX-2 DR1 could either serve as RARE or RXRE depending on cellular contexts.


Asunto(s)
Ciclooxigenasa 2/metabolismo , PPAR gamma/metabolismo , Tretinoina/metabolismo , Alitretinoína , Animales , Sitios de Unión , Línea Celular Tumoral , Ciclooxigenasa 2/genética , Activación Enzimática , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos ICR , Estructura Terciaria de Proteína
5.
Cell Biosci ; 11(1): 155, 2021 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-34348778

RESUMEN

BACKGROUND: Recently, we demonstrated that estrogen (E2) induces early growth response 1 (Egr1) to mediate its actions on the uterine epithelium by controlling progesterone receptor signaling for successful embryo implantation. EGR1 is a transcription factor that regulates the spectrum of target genes in many different tissues, including the uterus. E2-induced EGR1 regulates a set of genes involved in epithelial cell remodeling during embryo implantation in the uterus. However, only few target genes of EGR1 in the uterus have been identified. RESULT: The expression of ADAM metallopeptidase with thrombospondin type 1 motif 1 (Adamts-1) was significantly downregulated in the uteri of E2-treated ovariectomized (OVX) Egr1(-/-) mice. Immunostaining of ADAMTS-1 revealed its exclusive expression in the uterine epithelium of OVX wild-type but not Egr1(-/-) mice treated with E2. The expression profiles of Adamts-1 and Egr1 were similar in the uteri of E2-treated OVX mice at various time points tested. Pre-treatment with ICI 182, 780, a nuclear estrogen receptor (ER) antagonist, effectively inhibited the E2-dependent induction of Egr1 and Adamts-1. Pharmacologic inhibition of E2-induced ERK1/2 or p38 phosphorylation interfered with the induction of EGR1 and ADAMTS-1. Furthermore, ADAMTS-1, as well as EGR1, was induced in stroma cells surrounding the implanting blastocyst during embryo implantation. Transient transfection with EGR1 expression vectors significantly induced the expression of ADAMTS-1. Luciferase activity of the Adamts-1 promoter containing EGR1 binding sites (EBSs) was increased by EGR1 in a dose-dependent manner, suggesting functional regulation of Adamts-1 transcription by EGR1. Site-directed mutagenesis of EBS on the Adamts-1 promoter demonstrated that EGR1 directly binds to the EBS at -1151/-1134 among four putative EBSs. CONCLUSIONS: Collectively, we have demonstrated that Adamts-1 is a novel target gene of E2-ER-MAPK-EGR1, which is critical for embryo implantation in the mouse uterus during early pregnancy.

6.
Cells ; 10(7)2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34359838

RESUMEN

The open carrier system (OC) is used for vitrification due to its high efficiency in preserving female fertility, but concerns remain that it bears possible risks of cross-contamination. Closed carrier systems (CC) could be an alternative to the OC to increase safety. However, the viability and developmental competence of vitrified/warmed (VW) oocytes using the CC were significantly lower than with OC. We aimed to improve the efficiency of the CC. Metaphase II oocytes were collected from mice after superovulation and subjected to in vitro fertilization after vitrification/warming. Increasing the cooling/warming rate and exposure time to cryoprotectants as key parameters for the CC effectively improved the survival rate and developmental competence of VW oocytes. When all the conditions that improved the outcomes were applied to the conventional CC, hereafter named the modified vitrification/warming procedure using CC (mVW-CC), the viability and developmental competence of VW oocytes were significantly improved as compared to those of VW oocytes in the CC. Furthermore, mVW-CC increased the spindle normality of VW oocytes, as well as the cell number of blastocysts developed from VW oocytes. Collectively, our mVW-CC optimized for mouse oocytes can be utilized for humans without concerns regarding possible cross-contamination during vitrification in the future.


Asunto(s)
Blastocisto/citología , Criopreservación/métodos , Fertilización In Vitro/métodos , Oocitos/citología , Vitrificación , Animales , Biomarcadores/metabolismo , Blastocisto/metabolismo , Factor de Transcripción CDX2/genética , Factor de Transcripción CDX2/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Crioprotectores/farmacología , Dimetilsulfóxido/farmacología , Glicol de Etileno/farmacología , Femenino , Expresión Génica , Masculino , Metafase , Ratones , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Espermatozoides/fisiología , Sacarosa/farmacología
7.
Cells ; 10(6)2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34205802

RESUMEN

Advanced maternal age (AMA) is known to be related to the decrease in the quality and quantity of oocytes. Oocyte vitrification is now considered an established assisted reproductive technology for fertility preservation. However, it remains unclear whether the oocytes in older women are more sensitive to various insults during vitrification. Thus, we evaluated whether AMA affects cellular and molecular features and developmental outcomes of oocytes after vitrification in mice. The oocytes were grouped as young fresh (YF), young vitrified/warmed (YV), aged fresh (AF), and aged vitrified/warmed (AV). The survival rate of AV oocytes was significantly lower than that of YV oocytes. The rates of fertilization, cleavage, and blastocyst formation of AV oocytes were significantly lower than those of other groups. AV oocytes were represented as aberrations in mitochondria distribution, microvacuole size, and autophagosome formation, leading to delayed embryo development in mice. This delay was associated with a reduced number of total cells and trophectoderm in the blastocyst developed from AV oocytes. Collectively, AMA exaggerates the vulnerability of oocytes to cryo-damage that occurs during vitrification in mice, suggesting that the current vitrification protocols optimized for oocytes from young females should be modified for oocytes from aged women.


Asunto(s)
Blastocisto/metabolismo , Criopreservación , Desarrollo Embrionario , Edad Materna , Oocitos/metabolismo , Animales , Femenino , Fertilización In Vitro , Masculino , Ratones
8.
Cell Biosci ; 10(1): 132, 2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33292460

RESUMEN

BACKGROUND: Aberration of estrogen (E2) and/or progesterone (P4) signaling pathways affects expression of their target genes, which may lead to failure of embryo implantation and following pregnancy. Although many target genes of progesterone receptors (PRs) have been identified in uterine stroma, only a few PR targets have been reported in the epithelium. Secretory phospholipase A2-(PLA2)-X, a member of the PLA2 family that releases arachidonic acids for the synthesis of prostaglandins that are important for embryo implantation, is dysregulated in the endometrium of patients suffering from repeated implantation failure. However, it is not clear whether sPLA2-X is directly regulated by ovarian steroid hormones for embryo implantation in the uterus. RESULT: P4 induced the Pla2g10 encoding of secretory PLA2-X in the apical region of uterine LE of ovariectomized mice via PR in both time- and dose-dependent manners, whereas E2 significantly inhibited it. This finding is consistent with the higher expression of Pla2g10 at the diestrus stage, when P4 is elevated during the estrous cycle, and at P4-treated delayed implantation. The level of Pla2g10 on day 4 of pregnancy (day 4) was dramatically decreased on day 5, when PRs are absent in the LE. Luciferase assays of mutagenesis in uterine epithelial cells demonstrated that four putative PR response elements in a Pla2g10 promoter region are transcriptionally active for Pla2g10. Intrauterine delivery of small interfering RNA for Pla2g10 on day 3 significantly reduced the number of implantation sites, reinforcing the critical function(s) of Pla2g10 for uterine receptivity in mice. CONCLUSIONS: Pla2g10 is a novel PR target gene whose expression is exclusively localized in the apical region of the uterine LE for uterine receptivity for embryo implantation in mice.

9.
Steroids ; 115: 177-181, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27643453

RESUMEN

Two female sex steroid hormones, estrogen and progesterone, are crucial regulators of many physiological functions of reproductive organs. These two hormones are versatile factors linking growth, differentiation, metabolism, and death of cells in the uterus. In recent years, it has become evident that autophagy is involved in the effects of estrogen and progesterone on various cellular events in reproductive organs. Autophagy is the self-eating catabolic process which is linked to cell survival and death in many contexts. In this review, we focus on the new findings concerning the regulation of autophagic response by sex steroid hormones in responsive target organs. We also attempt to further expand our insight into intracellular signaling mediators governing this regulation.


Asunto(s)
Autofagia/fisiología , Estrógenos/metabolismo , Progesterona/metabolismo , Útero/metabolismo , Animales , Femenino , Humanos
10.
Sci Rep ; 6: 20242, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26833131

RESUMEN

DGCR8 is an RNA-binding protein that interacts with DROSHA to produce pre-microRNA in the nucleus, while DICER generates not only mature microRNA, but also endogenous small interfering RNAs in the cytoplasm. Here, we produced Dgcr8 conditional knock-out mice using progesterone receptor (PR)-Cre (Dgcr8(d/d)) and demonstrated that canonical microRNAs dependent on the DROSHA-DGCR8 complex are required for uterine development as well as female fertility in mice. Adult Dgcr8(d/d) females neither underwent regular reproductive cycles nor produced pups, whereas administration of exogenous gonadotropins induced normal ovulation in these mice. Interestingly, immune cells associated with acute inflammation aberrantly infiltrated into reproductive organs of pregnant Dgcr8(d/d) mice. Regarding uterine development, multiple uterine abnormalities were noticeable at 4 weeks of age when PR is significantly increased, and the severity of these deformities increased over time. Gland formation and myometrial layers were significantly reduced, and the stromal cell compartment did not expand and became atrophic during uterine development in these mice. These results were consistent with aberrantly reduced stromal cell proliferation and completely failed decidualization. Collectively, we suggest that DGCR8-dependent canonical microRNAs are essential for uterine development and physiological processes such as proper immune modulation, reproductive cycle, and steroid hormone responsiveness in mice.


Asunto(s)
Anomalías Múltiples/genética , Infertilidad/genética , MicroARNs/genética , Proteínas de Unión al ARN/genética , Anomalías Urogenitales/genética , Útero/anomalías , Anomalías Múltiples/metabolismo , Animales , Proliferación Celular , Cuerpo Lúteo , Modelos Animales de Enfermedad , Ciclo Estral/genética , Femenino , Regulación de la Expresión Génica , Hormonas Esteroides Gonadales/metabolismo , Infertilidad/metabolismo , Ratones , Oocitos/metabolismo , Especificidad de Órganos/genética , Organogénesis/genética , Embarazo , Proteínas de Unión al ARN/metabolismo , Células del Estroma/metabolismo , Anomalías Urogenitales/metabolismo , Útero/metabolismo
11.
Endocrinology ; 145(6): 2886-95, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15001550

RESUMEN

We previously showed that peroxisome proliferator-activated receptor delta (PPARdelta) is crucial for embryo implantation as a receptor for cyclooxygenase-2-derived prostacyclin in mice. PPARs belong to the nuclear receptor superfamily. They form heterodimer with a retinoid X receptor, recruit transcriptional cofactors, and bind to a specific recognition element for regulation of target genes. Although cofactors are generally shared by various nuclear receptors, some are involved in cell-specific events. The objective of this investigation was to examine interactions of transcriptional cofactors with PPARdelta in uterine cells for its effectiveness in regulating gene expression. We chose two uterine cellular systems: periimplantation mouse uterus and AN(3)CA human uterine cell line. As examined by in situ hybridization, steroid receptor coactivator (SRC)-2, SRC-3, PPAR-interacting protein, receptor-interacting protein 140 (RIP140), nuclear receptor corepressor (N-CoR), and silencing mediator for retinoid and thyroid hormone receptor (SMRT) exhibit overlapping expression with that of PPARdelta in the periimplantation mouse uterus. Glutathione-S-transferase (GST) pull-down assays show that PPARdelta physically interacts with SRC 1-3, RIP140, PPAR-binding protein, N-CoR, and SMRT in the absence of ligands, suggesting their potent interactions with PPARdelta. Transient transfection assays in AN(3)CA cells show that among members of the SRC family, only SRC-2 serves as a true coactivator for PPARdelta, whereas all SRC members could enhance PPARalpha-induced transcriptional activation. Interestingly, N-CoR and SMRT potently repress PPARdelta-induced transcriptional activation but fail to repress PPARalpha activity. RIP140 is effective in repressing basal and PPAR-induced transcriptional activation. Collectively, the results suggest that gene regulation by PPARdelta in the uterine cells uniquely responds to SRC-2, N-CoR, SMRT, or RIP140, and these interactions may be operative during implantation when these cofactors are abundantly expressed.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Útero/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Western Blotting , Línea Celular Tumoral , Proteínas de Unión al ADN/farmacología , Implantación del Embrión , Femenino , Humanos , Ratones , Ratones Endogámicos , Proteínas Nucleares/farmacología , Co-Represor 1 de Receptor Nuclear , Co-Represor 2 de Receptor Nuclear , Proteína de Interacción con Receptores Nucleares 1 , Receptores Citoplasmáticos y Nucleares/fisiología , Proteínas Represoras/farmacología , Factores de Tiempo , Distribución Tisular , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología , Útero/citología
12.
Int J Dev Biol ; 58(2-4): 183-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25023684

RESUMEN

Cells learn to thrive under unfavorable conditions by various mechanisms, and autophagy, self-eating, is one such mechanism. Autophagy is always ongoing in cells at a basal level to turn over old proteins, provide building blocks for new proteins, and to dispose of unnecessary byproducts of metabolism, and normally it does not cause deleterious effects on other parts of basic cellular processes. Autophagy is often dubbed a "double-edged sword", as it is a necessary process for many cells, but its exaggeration may lead to cell death. Evidence is accumulating that autophagy is crucially involved in specific aspects of reproduction. Several recent studies have illustrated how the uniqueness of self-eating is manifested in germ cells and embryos. In this review, we attempt to portray where this relatively young field of autophagy research is heading in the context of reproductive biology research.


Asunto(s)
Autofagia , Reproducción/fisiología , Animales , Humanos , Transducción de Señal
13.
Reprod Toxicol ; 50: 60-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25461906

RESUMEN

Coordinate actions of ovarian estrogen (E2) and progesterone (P4) via their own receptors are critical for establishing uterine receptivity for embryo implantation in the uterus. E2 regulates expression of an array of genes to mediate its major actions on heterogeneous uterine cell types. Here we have investigated regulatory mechanism(s) of E2 and bisphenol A (BPA), an endocrine disruptor with potent estrogenic activity on expression of early growth response 1 (Egr1), a zinc finger transcription factor that regulates cell growth, differentiation and apoptosis in the uterus. Egr1 was rapidly and transiently induced by E2 and BPA mainly in stromal cells via nuclear estrogen receptor (ER)-ERK1/2 pathway. ICI 182,780, an ER antagonist, effectively inhibited their actions on EGR1 expression following ERK1/2 phosphorylation. Administration of pharmacological inhibitors for ERK1/2, but not AKT significantly blocked EGR1 expression induced by E2 and BPA. P4 effectively dampened action(s) of E2 and BPA on Egr1 expression via nuclear progesterone receptor. Its antagonistic effects were partially interfered with RU486 pretreatment. Interestingly, EGR1 is specifically induced in stromal cells surrounding implanting blastocyst. Collectively, our results show that through nuclear ER-dependent ERK1/2 phosphorylation, not only E2 but also endocrine disruptors with estrogenic activity such as BPA rapidly and transiently induce Egr1 which may be important for embryo implantation and decidualization in mouse uterus.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Estrógenos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Fenoles/farmacología , Receptores de Estrógenos/fisiología , Útero/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Proteína 3 de la Respuesta de Crecimiento Precoz/fisiología , Femenino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Progesterona/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Útero/efectos de los fármacos
14.
Clin Exp Reprod Med ; 38(1): 1-5, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22384410

RESUMEN

The maturation process of mammalian oocytes accompanies an extensive rearrangement of the cytoskeleton and associated proteins. As this process requires a delicate interplay between the cytoskeleton and its regulators, it is often targeted by various external and internal adversaries that affect the congression and/or segregation of chromosomes. Asymmetric cell division in oocytes also requires specific regulators of the cytoskeleton, including formin-2 and small GTPases. Recent literature providing clues regarding how actin filaments and microtubules interact during spindle migration in mouse oocytes are highlighted in this review.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA