Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 13(7)2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38607081

RESUMEN

Increased activation of ovarian primordial follicles in Erß knockout (ErßKO) rats becomes evident as early as postnatal day 8.5. To identify the ERß-regulated genes that may control ovarian primordial follicle activation, we analyzed the transcriptome profiles of ErßKO rat ovaries collected on postnatal days 4.5, 6.5, and 8.5. Compared to wildtype ovaries, ErßKO ovaries displayed dramatic downregulation of Indian hedgehog (Ihh) expression. IHH-regulated genes, including Hhip, Gli1, and Ptch1, were also downregulated in ErßKO ovaries. This was associated with a downregulation of steroidogenic enzymes Cyp11a1, Cyp19a1, and Hsd17b1. The expression of Ihh remained very low in ErßKO ovaries despite the high levels of Gdf9 and Bmp15, which are known upregulators of Ihh expression in the granulosa cells of activated ovarian follicles. Strikingly, the downregulation of the Ihh gene in ErßKO ovaries began to disappear on postnatal day 16.5 and recovered on postnatal day 21.5. In rat ovaries, the first wave of primordial follicles is rapidly activated after their formation, whereas the second wave of primordial follicles remains dormant in the ovarian cortex and slowly starts activating after postnatal day 12.5. We localized the expression of Ihh mRNA in postnatal day 8.5 wildtype rat ovaries but not in the age-matched ErßKO ovaries. In postnatal day 21.5 ErßKO rat ovaries, we detected Ihh mRNA mainly in the activated follicles in the ovaries' peripheral regions. Our findings indicate that the expression of Ihh in the granulosa cells of the activated first wave of ovarian follicles depends on ERß.


Asunto(s)
Receptor beta de Estrógeno , Proteínas Hedgehog , Animales , Femenino , Ratas , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Folículo Ovárico/metabolismo , Ovario/metabolismo , ARN Mensajero/metabolismo
2.
Int J Mol Sci ; 25(6)2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38542176

RESUMEN

Loss of ERß increases primordial follicle growth activation (PFGA), leading to premature ovarian follicle reserve depletion. We determined the expression and gene regulatory functions of ERß in dormant primordial follicles (PdFs) and activated primary follicles (PrFs) using mouse models. PdFs and PrFs were isolated from 3-week-old Erß knockout (Erßnull) mouse ovaries, and their transcriptomes were compared with those of control Erßfl/fl mice. We observed a significant (≥2-fold change; FDR p-value ≤ 0.05) deregulation of approximately 5% of genes (866 out of 16,940 genes, TPM ≥ 5) in Erßnull PdFs; ~60% (521 out of 866) of the differentially expressed genes (DEGs) were upregulated, and 40% were downregulated, indicating that ERß has both transcriptional enhancing as well as repressing roles in dormant PdFs. Such deregulation of genes may make the Erßnull PdFs more susceptible to increased PFGA. When the PdFs undergo PFGA and form PrFs, many new genes are activated. During PFGA of Erßfl/fl follicles, we detected a differential expression of ~24% genes (4909 out of 20,743; ≥2-fold change; FDR p-value ≤ 0.05; TPM ≥ 5); 56% upregulated and 44% downregulated, indicating the gene enhancing and repressing roles of Erß-activated PrFs. In contrast, we detected a differential expression of only 824 genes in Erßnull follicles during PFGA (≥2-fold change; FDR p-value ≤ 0.05; TPM ≥ 5). Moreover, most (~93%; 770 out of 824) of these DEGs in activated Erßnull PrFs were downregulated. Such deregulation of genes in Erßnull activated follicles may impair their inhibitory role on PFGA. Notably, in both Erßnull PdFs and PrFs, we detected a significant number of epigenetic regulators and transcription factors to be differentially expressed, which suggests that lack of ERß either directly or indirectly deregulates the gene expression in PdFs and PrFs, leading to increased PFGA.


Asunto(s)
Receptor beta de Estrógeno , Folículo Ovárico , Femenino , Ratones , Animales , Receptor beta de Estrógeno/metabolismo , Folículo Ovárico/metabolismo , Ovario/metabolismo , Regulación de la Expresión Génica , Transcriptoma , Ratones Noqueados
3.
Int J Mol Sci ; 24(17)2023 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-37685885

RESUMEN

Ovarian follicles undergo a series of dynamic changes following the ovulatory surge of luteinizing hormone including cumulus expansion, oocyte maturation, ovulation, and luteinization. Post-transcriptional gene regulatory events are critical for mediating LH follicular responses, and among all RNA isoforms, circular RNA (circRNA) is one of the most abundant forms present in cells, yet they remain the least studied. Functionally, circRNA can act as miRNA sponges, protein sponges/decoys, and regulators of transcription and translation. In the context of ovarian follicular development, the identity and roles of circRNA are relatively unknown. In the present study, high throughput RNA sequencing of granulosa cells immediately prior to and 4-h after the LH/hCG surge identified 42,381 circRNA originating from 7712 genes. A total of 54 circRNA were identified as differentially expressed between 0-h and 4-h time points (Fold Change ± 1.5, FDR ≤ 0.1), among them 42 circRNA were upregulated and 12 circRNA were downregulated. All differentially expressed circRNA between the 0-h and 4-h groups were subjected to circinteractome analysis and identified networks of circRNA-protein and circRNA-miRNA were further subjected to "micro-RNA target filter analysis" in Ingenuity Pathway Analyses, which resulted in the identification of miRNA targeted mRNAs. A comparison of these circRNA target mRNAs with LH-induced mRNAs identified Runx2, Egfr, Areg, Sult1el, Cyp19a1, Cyp11a1, and Hsd17b1 as targets of circKif2, circVcan, circMast4, and circMIIt10. These newly identified LH/hCG-induced circRNA, their target miRNA and protein networks provide new insights into the complex interactions associated with periovulatory follicular development.


Asunto(s)
Células de la Granulosa , ARN Circular , Femenino , Animales , Ratones , ARN Circular/genética , Folículo Ovárico , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol , Citocromo P-450 CYP1A1
4.
Int J Mol Sci ; 23(22)2022 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-36430478

RESUMEN

Adenosine deaminases acting on RNA-(ADAR) comprise one family of RNA editing enzymes that specifically catalyze adenosine to inosine (A-to-I) editing. A granulosa cell (GC) specific Adar depleted mouse model [Adar flox/flox:Cyp19a1-Cre/+ (gcAdarKO)] was used to evaluate the role of ADAR1 during the periovulatory period. Loss of Adar in GCs led to failure to ovulate at 16 h post-hCG, delayed oocyte germinal vesicle breakdown and severe infertility. RNAseq analysis of GC collected from gcAdarKO and littermate control mice at 0 and 4 h post-hCG following a super-ovulatory dose of eCG (48 h), revealed minimal differences after eCG treatment alone (0 h), consistent with normal folliculogenesis observed histologically and uterine estrogenic responses. In contrast, 300 differential expressed genes (DEGs; >1.5-fold change and FDRP < 0.1) were altered at 4 h post-hCG. Ingenuity pathway analysis identified many downstream targets of estrogen and progesterone pathways, while multiple genes involved in inflammatory responses were upregulated in the gcAdarKO GCs. Temporal expression analysis of GCs at 0, 4, 8, and 12 h post-hCG of Ifi44, Ifit1, Ifit3b, and Oas1g and Ovgp1 confirmed upregulation of these inflammatory and interferon genes and downregulation of Ovgp1 a glycoprotein involved in oocyte zona pellucida stability. Thus, loss of ADAR1 in GCs leads to increased expression of inflammatory and interferon response genes which are temporally linked to ovulation failure, alterations in oocyte developmental progression and infertility.


Asunto(s)
Infertilidad , Ovulación , Femenino , Animales , Ratones , Ovulación/genética , Células de la Granulosa , Interferones , Infertilidad/genética , Oocitos , Adenosina
5.
Front Endocrinol (Lausanne) ; 13: 917464, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36072937

RESUMEN

Kisspeptins (KPs) secreted from the hypothalamic KP neurons act on KP receptors (KPRs) in gonadotropin (GPN) releasing hormone (GnRH) neurons to produce GnRH. GnRH acts on pituitary gonadotrophs to induce secretion of GPNs, namely follicle stimulating hormone (FSH) and luteinizing hormone (LH), which are essential for ovarian follicle development, oocyte maturation and ovulation. Thus, hypothalamic KPs regulate oocyte maturation indirectly through GPNs. KPs and KPRs are also expressed in the ovarian follicles across species. Recent studies demonstrated that intraovarian KPs also act directly on the KPRs expressed in oocytes to promote oocyte maturation and ovulation. In this review article, we have summarized published reports on the role of hypothalamic and ovarian KP-signaling in oocyte maturation. Gonadal steroid hormones regulate KP secretion from hypothalamic KP neurons, which in turn induces GPN secretion from the hypothalamic-pituitary (HP) axis. On the other hand, GPNs secreted from the HP axis act on the granulosa cells (GCs) and upregulate the expression of ovarian KPs. While KPs are expressed predominantly in the GCs, the KPRs are in the oocytes. Expression of KPs in the ovaries increases with the progression of the estrous cycle and peaks during the preovulatory GPN surge. Intrafollicular KP levels in the ovaries rise with the advancement of developmental stages. Moreover, loss of KPRs in oocytes in mice leads to failure of oocyte maturation and ovulation similar to that of premature ovarian insufficiency (POI). These findings suggest that GC-derived KPs may act on the KPRs in oocytes during their preovulatory maturation. In addition to the intraovarian role of KP-signaling in oocyte maturation, in vivo, a direct role of KP has been identified during in vitro maturation of sheep, porcine, and rat oocytes. KP-stimulation of rat oocytes, in vitro, resulted in Ca2+ release and activation of the mitogen-activated protein kinase, extracellular signal-regulated kinase 1 and 2. In vitro treatment of rat or porcine oocytes with KPs upregulated messenger RNA levels of the factors that favor oocyte maturation. In clinical trials, human KP-54 has also been administered successfully to patients undergoing assisted reproductive technologies (ARTs) for increasing oocyte maturation. Exogenous KPs can induce GPN secretion from hypothalamus; however, the possibility of direct KP action on the oocytes cannot be excluded. Understanding the direct in vivo and in vitro roles of KP-signaling in oocyte maturation will help in developing novel KP-based ARTs.


Asunto(s)
Kisspeptinas , Oogénesis , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Ratones , Oocitos/fisiología , Ratas , Ovinos , Porcinos
6.
Front Genet ; 13: 828086, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35401699

RESUMEN

DOT1L is essential for embryonic hematopoiesis but the precise mechanisms of its action remain unclear. The only recognized function of DOT1L is histone H3 lysine 79 (H3K79) methylation, which has been implicated in both transcriptional activation and repression. We observed that deletion of the mouse Dot1L gene (Dot1L-KO) or selective mutation of its methyltransferase domain (Dot1L-MM) can differentially affect early embryonic erythropoiesis. However, both mutations result in embryonic lethality by mid-gestation and growth of hematopoietic progenitor cells (HPCs) is similarly affected in extensively self-renewing erythroblast (ESRE) cultures established from yolk sac cells. To understand DOT1L-mediated gene regulation and to clarify the role of H3K79 methylation, we analyzed whole transcriptomes of wildtype and Dot1L-mutant ESRE cells. We observed that more than 80% of the differentially expressed genes (DEGs) were upregulated in the mutant ESRE cells either lacking the DOT1L protein or the DOT1L methyltransferase activity. However, approximately 45% of the DEGs were unique to either mutant group, indicating that DOT1L possesses both methyltransferase-dependent and -independent gene regulatory functions. Analyses of Gene Ontology and signaling pathways for the DEGs were consistent, with DEGs that were found to be common or unique to either mutant group. Genes related to proliferation of HPCs were primarily impacted in Dot1L-KO cells, while genes related to HPC development were affected in the Dot1L-MM cells. A subset of genes related to differentiation of HPCs were affected in both mutant groups of ESREs. Our findings suggest that DOT1L primarily acts to repress gene expression in HPCs, and this function can be independent of its methyltransferase activity.

7.
Front Cell Dev Biol ; 10: 918235, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36589740

RESUMEN

SATB homeobox proteins are important regulators of developmental gene expression. Among the stem cell lineages that emerge during early embryonic development, trophoblast stem (TS) cells exhibit robust SATB expression. Both SATB1 and SATB2 act to maintain the trophoblast stem-state. However, the molecular mechanisms that regulate TS-specific Satb expression are not yet known. We identified Satb1 variant 2 as the predominant transcript in trophoblasts. Histone marks, and RNA polymerase II occupancy in TS cells indicated an active state of the promoter. A novel cis-regulatory region with active histone marks was identified ∼21 kbp upstream of the variant 2 promoter. CRISPR/Cas9 mediated disruption of this sequence decreased Satb1 expression in TS cells and chromosome conformation capture analysis confirmed looping of this distant regulatory region into the proximal promoter. Scanning position weight matrices across the enhancer predicted two ELF5 binding sites in close proximity to SATB1 sites, which were confirmed by chromatin immunoprecipitation. Knockdown of ELF5 downregulated Satb1 expression in TS cells and overexpression of ELF5 increased the enhancer-reporter activity. Interestingly, ELF5 interacts with SATB1 in TS cells, and the enhancer activity was upregulated following SATB overexpression. Our findings indicate that trophoblast-specific Satb1 expression is regulated by long-range chromatin looping of an enhancer that interacts with ELF5 and SATB proteins.

8.
Int J Mol Sci ; 22(19)2021 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-34638689

RESUMEN

Gonadotropins are essential for regulating ovarian development, steroidogenesis, and gametogenesis. While follicle stimulating hormone (FSH) promotes the development of ovarian follicles, luteinizing hormone (LH) regulates preovulatory maturation of oocytes, ovulation, and formation of corpus luteum. Cognate receptors of FSH and LH are G-protein coupled receptors that predominantly signal through cAMP-dependent and cAMP-independent mechanisms that activate protein kinases. Subsequent vital steps in response to gonadotropins are mediated through activation or inhibition of transcription factors required for follicular gene expression. Estrogen receptors, classical ligand-activated transcriptional regulators, play crucial roles in regulating gonadotropin secretion from the hypothalamic-pituitary axis as well as gonadotropin function in the target organs. In this review, we discuss the role of estrogen receptor ß (ERß) regulating gonadotropin response during folliculogenesis. Ovarian follicles in Erß knockout (ErßKO) mutant female mice and rats cannot develop beyond the antral state, lack oocyte maturation, and fail to ovulate. Theca cells (TCs) in ovarian follicles express LH receptor, whereas granulosa cells (GCs) express both FSH receptor (FSHR) and LH receptor (LHCGR). As oocytes do not express the gonadotropin receptors, the somatic cells play a crucial role during gonadotropin induced oocyte maturation. Somatic cells also express high levels of estrogen receptors; while TCs express ERα and are involved in steroidogenesis, GCs express ERß and are involved in both steroidogenesis and folliculogenesis. GCs are the primary site of ERß-regulated gene expression. We observed that a subset of gonadotropin-induced genes in GCs, which are essential for ovarian follicle development, oocyte maturation and ovulation, are dependent on ERß. Thus, ERß plays a vital role in regulating the gonadotropin responses in ovary.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Receptor beta de Estrógeno/metabolismo , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/metabolismo , Células Tecales/metabolismo , Animales , Gonadotropina Coriónica/genética , Receptor beta de Estrógeno/genética , Femenino , Hormona Folículo Estimulante/genética , Humanos , Ratones , Ratones Noqueados , Ratas
9.
Mol Cell Endocrinol ; 528: 111212, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33676987

RESUMEN

Follicle development beyond the preantral stage is dependent on gonadotropins. FSH signaling is crucial for the advancement of preantral follicles to the antral stage, and LH signaling is essential for further maturation of preovulatory follicles. Estrogen is intricately tied to gonadotropin signaling during the advanced stages of folliculogenesis. We observed that Erßnull ovarian follicles fail to develop beyond the antral stage, even after exogenous gonadotropin stimulation. As ERß is primarily expressed in the granulosa cells (GCs), we explored the gonadotropin-regulated GC genes that induce maturation of antral follicles. Synchronized follicle development was induced by administration of exogenous gonadotropins to wildtype 4-wk-old female rats. The GC transcriptome was analyzed via RNA-sequencing before and after gonadotropin stimulation. An Erßnull mutant model that fails to show follicle maturation was also included in order to identify the ERß-regulated genes involved at this step. We observed that specific groups of genes were differentially expressed in response to PMSG or hCG administration in wildtype rats. While some of the PMSG or hCG-induced genes showed a similar expression pattern in Erßnull GCs, a subset of PMSG- or hCG-induced genes showed a differential expression pattern in Erßnull GCs. These latter ERß-regulated genes included previously known FSH or LH target genes including Lhcgr, Cyp11a1, Cyp19a1, Pgr, Runx2, Egfr, Kiss1, and Ptgs2, which are involved in follicle development, oocyte maturation, and ovulation. We also identified novel ERß-regulated genes including Jaml, Galnt6, Znf750, Dusp9, Wnt16, and Mageb16 that failed to respond to gonadotropin stimulation in Erßnull GCs. Our findings indicate that the gonadotropin-induced spatiotemporal pattern of gene expression is essential for ovarian follicle maturation beyond the antral stage. However, expression of a subset of those gonadotropin-induced genes is dependent on transcriptional regulation by ERß.


Asunto(s)
Gonadotropina Coriónica/administración & dosificación , Receptor beta de Estrógeno/genética , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes/efectos de los fármacos , Folículo Ovárico/crecimiento & desarrollo , Animales , Gonadotropina Coriónica/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/química , Células de la Granulosa/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Mutación con Pérdida de Función , Folículo Ovárico/química , Folículo Ovárico/efectos de los fármacos , Ratas , Análisis de Secuencia de ARN
10.
Mol Cell Endocrinol ; 527: 111208, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33592287

RESUMEN

Kisspeptin (KISS1) signaling in the hypothalamic-pituitary (H-P) axis plays an essential role in regulating gonadotropin secretion. KISS1 and KISS1 receptor (KISS1R) are also expressed in the ovary; however, the role of intraovarian KISS1 signaling remains unclear. Granulosa cell (GC)-specific expression of KISS1, and oocyte-specific expression of KISS1R indicate that GC-derived KISS1 may act on oocytes. Expression of KISS1 in GCs is induced by gonadotropins but it is absent in estrogen receptor ß knockout (Erßnull) rat ovaries. We also observed that gonadotropin stimulation failed to induce maturation of Erßnull oocytes. Interestingly, KISS1 treatment of cumulus oocyte complexes (COCs) isolated from antral follicles promotes in vitro maturation of oocytes. Treatment of oocytes with KISS1 induced intracellular Ca2+ release, and increased activation of MAP kinase ERK1/2. KISS1 treatment also induced the expression of oocyte genes that are crucial for differentiation of GCs, and maturation of oocytes. Our findings suggest that ovarian KISS1-signaling plays an important role in gonadotropin induced follicle development and oocyte maturation.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Células de la Granulosa/metabolismo , Kisspeptinas/metabolismo , Sistema de Señalización de MAP Quinasas , Oocitos/metabolismo , Animales , Receptor beta de Estrógeno/genética , Femenino , Kisspeptinas/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas
11.
Front Cell Dev Biol ; 9: 813503, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35111761

RESUMEN

DOT1-like (DOT1L) histone methyltransferase is essential for mammalian erythropoiesis. Loss of DOT1L in knockout (Dot1l-KO) mouse embryos resulted in lethal anemia at midgestational age. The only recognized molecular function of DOT1L is its methylation of histone H3 lysine 79 (H3K79). We generated a Dot1l methyltransferase mutant (Dot1l-MM) mouse model to determine the role of DOT1L methyltransferase activity in early embryonic hematopoiesis. Dot1l-MM embryos failed to survive beyond embryonic day 13.5 (E13.5), similarly to Dot1l-KO mice. However, when examined at E10.5, Dot1l-MM embryos did not exhibit overt anemia like the Dot1l-KO. Vascularity and the presence of red blood cells in the Dot1l-MM yolk sacs as well as in the AGM region of Dot1l-MM embryos appeared to be similar to that of wildtype. In ex vivo cultures of yolk sac cells, Dot1l-MM primitive erythroblasts formed colonies comparable to those of the wildtype. Although ex vivo cultures of Dot1l-MM definitive erythroblasts formed relatively smaller colonies, inhibition of DOT1L methyltransferase activity in vivo by administration of EPZ-5676 minimally affected the erythropoiesis. Our results indicate that early embryonic erythropoiesis in mammals requires a DOT1L function that is independent of its intrinsic methyltransferase activity.

12.
Data Brief ; 30: 105562, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32368594

RESUMEN

Tg26 mice are robust models of human immunodeficiency virus 1 associated nephropathy (HIVAN). These mice are useful for HIVAN pathology analysis, and recent studies suggest that the Tg26 mouse model is an excellent model of other chronic kidney diseases. We performed RNA seq analysis of differential gene expression in the kidneys of Tg26 mice. Kidneys were collected from Tg26 mice and wildtype (WT) littermates at 3 months of age. The raw data were analyzed for differential gene expression using a negative binomial generalized linear model in the DeSeq2 software package. We used P-Value ≤0.05 and an absolute fold change of 1.5 to identify top 50 upregulated and top 50 downregulated differentially expressed genes between the WT and Tg26 mice. As expected inflammatory genes were among the top differentially regulated genes. Our data provides yet another level of information to help gain a more comprehensive understanding of disease progression and identify potential drug targets for HIVAN and chronic kidney diseases.

13.
Data Brief ; 30: 105405, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32280735

RESUMEN

Disruption of estrogen receptor beta (ESR2) dysregulates granulosa cell genes essential for follicle maturation and ovulation. The datasets presented in this article depict gonadotropin-induced genes, which are differentially expressed in Esr2-null rat granulosa cells. Synchronized follicle development was initiated in four-week-old wildtype and Esr2-null female rats by administration of PMSG. Forty-eight hours after PMSG injection, further maturation of ovarian follicles was induced by hCG treatment. Granulosa cells were collected from the ovaries before gonadotropin administration, 48 h after PMSG treatment, and 4 h after hCG injection to the PMSG-treated rats. Total RNA was purified from granulosa cells and whole transcriptome was assessed by RNA-sequencing on an Illumina HiSeq X platform. RNA-seq data of wildtype and Esr2-null granulosa cells were analyzed and differentially expressed genes were identified by CLC Genomics Workbench. Gonadotropin-induced genes were identified by comparing the transcriptome data of PMSG- or hCG-induced wildtype granulosa cells with those without gonadotropin treatment. Furthermore, differentially expressed genes in Esr2-null granulosa cells were determined by comparing the transcriptome data with that of wildtype granulosa cells. These datasets can be used to recognize the gonadotropin-induced genes in granulosa cells that are Esr2-regulated and important for ovarian follicle maturation.

14.
Endocrinology ; 161(4)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32141511

RESUMEN

Over the entire reproductive lifespan in mammals, a fixed number of primordial follicles serve as the source of mature oocytes. Uncontrolled and excessive activation of primordial follicles can lead to depletion of the ovarian reserve. We observed that disruption of estrogen receptor ß (ESR2) signaling results in increased activation of primordial follicles in Esr2-null (Esr2-/-) rats. However, follicle assembly was unaffected, and the total number of follicles remained comparable between neonatal wild-type and Esr2-/- ovaries. While the activated follicle counts were increased in Esr2-/- ovary, the number of primordial follicles were markedly decreased. Excessive recruitment of primordial follicles led to premature ovarian senescence in Esr2-/- rats and was associated with reduced levels of serum AMH and estradiol. Disruption of ESR2 signaling through administration of a selective antagonist (PHTPP) increased the number of activated follicles in wildtype rats, whereas a selective agonist (DPN) decreased follicle activation. In contrast, primordial follicle activation was not increased in the absence of ESR1, indicating that the regulation of primordial follicle activation is ESR2 specific. Follicle activation was also increased in Esr2 mutants lacking the DNA binding domain, suggesting a role for the canonical transcriptional activation function. Both primordial and activated follicles express ESR2, suggesting a direct regulatory role for ESR2 within these follicles. We also detected that loss of ESR2 augmented the activation of AKT, ERK, and mTOR pathways. Our results indicate that the lack of ESR2 upregulated both granulosa and oocyte factors, which can facilitate AKT and mTOR activation in Esr2-/- ovaries leading to increased activation of primordial follicles.


Asunto(s)
Hormona Antimülleriana/sangre , Estradiol/sangre , Receptor beta de Estrógeno/genética , Folículo Ovárico/metabolismo , Reserva Ovárica/fisiología , Animales , Moduladores de los Receptores de Estrógeno/farmacología , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/metabolismo , Femenino , Diana Mecanicista del Complejo 1 de la Rapamicina , Nitrilos/farmacología , Folículo Ovárico/efectos de los fármacos , Reserva Ovárica/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Transducción de Señal/efectos de los fármacos
15.
Data Brief ; 27: 104786, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31788515

RESUMEN

Disruption of estrogen receptor beta (ESR2) dysregulates oocyte maturation, which leads to failure of ovulation. We investigated ESR2-regulated genes during gonadotropin-induced oocyte maturation using RNA-sequencing. Through the administration of pregnant mare's serum gonadotropin (PMSG), synchronized follicle development was initiated in four-week-old wildtype and Esr2-null female rats. Forty-eight hours after the PMSG injection, human chorionic gonadotropin (hCG) was used for further maturation. Oocytes were collected from the ovaries 4 h after hCG injection. The total RNA was isolated from the oocytes and the whole oocyte transcriptome was determined by RNA-sequencing on the Illumina HiSeq4000 sequencer. RNA-sequencing data of wildtype and Esr2-null oocytes were analyzed, and differentially expressed genes were identified using the CLC Genomics Workbench. Whole oocyte transcriptome data of wildtype and Esr2-null oocytes were compared to identify the differentially expressed genes. Raw data are deposited to the NCBI Sequence Read Archive (SRA) and analyzed data are presented in this data article. These datasets can be utilized to identify the gonadotropin-induced genes in oocytes that are ESR2-regulated and important to oocyte maturation.

16.
Data Brief ; 27: 104749, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31886333

RESUMEN

SATB homeobox 1 (SATB1) and its heterodimeric partner SATB2 play an important regulatory role in maintaining proliferation of trophoblast stem (TS) cells and in inhibiting trophoblast differentiation. To identify the SATB-regulated genes in TS cells, we studied the transcriptome changes in a 'loss of function' model of Rcho-1 rat TS cell line. Satb1 gene expression was silenced by lentiviral delivery of shRNAs targeted to exon 9 and exon 12. An Egfp shRNA was used as a non-targeted control. Total RNA was purified from shRNA-transduced Rcho-1 cells, and whole transcriptome was assessed by RNA-sequencing on an Illumina HiSeq X platform. Differentially expressed genes in Satb1 shRNA-transduced cells were identified by analyses of the RNA-sequencing data using CLC Genomics Workbench. Differentially expressed genes with each of the two different shRNAs were compared to identify SATB1-target genes and to eliminate the potential off-targets of the shRNAs. These datasets can be used to identify the SATB-regulated genes in TS cells and to understand the molecular mechanisms that regulate trophoblast proliferation and inhibit differentiation.

17.
Dis Model Mech ; 12(12)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31727625

RESUMEN

Notch pathway activation plays a central role in the pathogenesis of many glomerular diseases. We have previously shown that Notch4 expression was upregulated in various renal cells in human immunodeficiency virus (HIV)-associated nephropathy (HIVAN) patients and rodent models of HIVAN. In this study, we examined whether the Notch pathway can be distinctly activated by HIV-1 gene products and whether Notch4, in particular, can influence disease progression. Using luciferase reporter assays, we did not observe activation of the NOTCH4 promoter with the HIV protein Nef in podocytes. Further, we observed upregulated expression of a gamma secretase complex protein, presenilin 1, but not Notch4, in podocytes infected with an HIV-1 expression construct. To assess the effects of Notch4 on HIVAN disease progression, we engineered Tg26 mice with global deletion of the Notch4 intracellular domain (Notch4dl ), which is required for signaling function. These mice (Notch4d1/Tg26+ ) showed a significant improvement in renal function and a significant decrease in mortality compared to Tg26 mice. Histological examination of kidneys showed that Notch4d1/Tg26+ mice had overall glomerular, tubulointerstitial injury and a marked decrease in interstitial inflammation. A significant decrease in the proliferating cells was observed in the tubulointerstitial compartments of Notch4d1/Tg26+ mice. Consistent with the diminished inflammation, kidneys from Notch4d1/Tg26+ mice also showed a significant decrease in expression of the inflammatory cytokine transcripts Il-6 and Ccl2, as well as the master inflammatory transcription factor NF-κB (Nfkb1 transcripts and p65 protein). These data identify Notch4 as an important mediator of tubulointerstitial injury and inflammation in HIVAN and a potential therapeutic target.


Asunto(s)
Nefropatía Asociada a SIDA/metabolismo , Inflamación/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Receptor Notch4/metabolismo , Animales , Proliferación Celular , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Eliminación de Gen , Células HEK293 , Humanos , Riñón/metabolismo , Masculino , Ratones , Ratones Transgénicos , Podocitos/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Resultado del Tratamiento , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
18.
Theriogenology ; 136: 1-6, 2019 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-31229779

RESUMEN

The objective of this study was to test the hypothesis that Leptin induced in vitro growth in preantral follicles in sheep involves modulation of P450 aromatase expression and steroidogenesis. Accordingly, the expression of P450 aromatase gene was studied in the cumulus cells and oocytes isolated from different stages of preantral follicles (PFs') grown in vivo, cultured in TCM 199B, TCM 199B + Leptin (10 ng/ml) (TCM199BL) or a standard PF culture medium supplemented with Leptin (10 ng/ml) (SML). Ovarian follicles grown in vivo or in SML expressed P450 aromatase both in cumulus cells and oocytes at all the development stages. In the oocytes from PFs' grown in vitro, P450 expression was consistently lower than in those from in vivo grown follicles at all except the preantral stage. The patterns of expression of aromatase gene in the cumulus cells from in vivo grown and the PFs' cultured in TCM 199BL were similar. Significantly higher levels of progesterone production were supported by SML at all the development stages than the other two media. Oestradiol concentration in the spent TCM 199B and SML showed a significant increase as the development progressed from preantral to large antral stage. However, such increase was not sustained beyond early antral stage in the PFs' cultured in TCM199BL. It is concluded that Leptin modulates the expression P450 aromatase while supporting the in vitro development of the ovarian follicles in sheep.


Asunto(s)
Aromatasa/metabolismo , Estrógenos/biosíntesis , Leptina/farmacología , Folículo Ovárico/efectos de los fármacos , Progesterona/biosíntesis , Ovinos , Animales , Aromatasa/genética , Medios de Cultivo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Técnicas de Cultivo de Tejidos/veterinaria
19.
Mol Cell Endocrinol ; 490: 47-56, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30974146

RESUMEN

The liver helps maintain energy homeostasis by synthesizing and storing glucose and lipids. Gonadal steroids, particularly estrogens, play an important role in regulating metabolism. As estrogens are considered female hormones, metabolic disorders related to the disruption of estrogen signaling have mostly been studied in females. Estrogen receptor alpha (ESR1) is the predominant receptor in both the male and female liver, and it mediates the hepatic response to estrogens. Loss of ESR1 increases weight gain and obesity in female rats, while reducing the normal growth in males. Although Esr1-/- male rats have a reduced body weight, they exhibit increased adipose deposition and impaired glucose tolerance. We further investigated whether these metabolic disorders in Esr1-/- male rats were linked with the loss of transcriptional regulation by ESR1 in the liver. To identify the ESR-regulated genes, RNA-sequencing was performed on liver mRNAs from wildtype and Esr1-/- male rats. Based on an absolute fold change of ≥2 with a p-value ≤ 0.05, a total of 706 differentially expressed genes were identified in the Esr1-/- male liver: 478 downregulated, and 228 upregulated. Pathway analyses demonstrate that the differentially expressed genes include transcriptional regulators (Cry1, Nr1d1, Nr0b2), transporters (Slc1a2), and regulators of biosynthesis (Cyp7b1, Cyp8b1), and hormone metabolism (Hsd17b2, Sult1e1). Many of these genes are also integral parts of the lipid and carbohydrate metabolism pathways in the liver. Interestingly, certain critical regulators of the metabolic pathways displayed a sexual dimorphism in expression, which may explain the divergent weight gain in Esr1-/- male and female rats despite common metabolic dysfunctions.


Asunto(s)
Metabolismo de los Hidratos de Carbono/genética , Receptor alfa de Estrógeno/metabolismo , Regulación de la Expresión Génica , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Adiposidad , Animales , Femenino , Ontología de Genes , Glucosa/metabolismo , Insulina/metabolismo , Lípidos/sangre , Masculino , Modelos Biológicos , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Aumento de Peso
20.
Data Brief ; 24: 103826, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31016213

RESUMEN

Estrogen signaling plays an important role in the pathophysiology of prostatic hyperplasia. While signaling through estrogen receptor alpha (ESR1) increases proliferation of stromal cells, estrogen receptor beta (ESR2) plays an anti-proliferative and differentiating role in glandular epithelium. Disruption of ESR2 signaling resulted in prostatic glandular hyperplasia in the rat. To identify the ESR2-target genes, and the molecular mechanisms involved, we performed RNA-seq analyses in prostate glands of Esr2 knockout (Esr2-/-) and age matched wildtype rats. The raw data were analyzed using CLC genomics workbench. High quality RNA-seq reads were aligned to the Rattus norvegicus genome. Differentially expressed genes were identified based on an absolute fold change of 2 with pValue ≤0.05. Of the total 32,623 genes detected, 824 were differentially expressed in Esr2-/- prostate glands, 550 downregulated and 274 upregulated. Pathway analyses identified altered expression of genes involved in epithelial proliferation and benign tumor formation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...