Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Reprod Dev ; 91(3): e23739, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38480999

RESUMEN

During male fetal development, testosterone plays an essential role in the differentiation and maturation of the male reproductive system. Deficient fetal testosterone production can result in variations of sex differentiation that may cause infertility and even increased tumor incidence later in life. Fetal Leydig cells in the fetal testis are the major androgen source in mammals. Although fetal and adult Leydig cells are similar in their functions, they are two distinct cell types, and therefore, the knowledge of adult Leydig cells cannot be directly applied to understanding fetal Leydig cells. This review summarizes our current knowledge of fetal Leydig cells regarding their cell biology, developmental biology, and androgen production regulation in rodents and human. Fetal Leydig cells are present in basement membrane-enclosed clusters in between testis cords. They originate from the mesonephros mesenchyme and the coelomic epithelium and start to differentiate upon receiving a Desert Hedgehog signal from Sertoli cells or being released from a NOTCH signal from endothelial cells. Mature fetal Leydig cells produce androgens. Human fetal Leydig cell steroidogenesis is LHCGR (Luteinizing Hormone Chronic Gonadotropin Receptor) dependent, while rodents are not, although other Gαs -protein coupled receptors might be involved in rodent steroidogenesis regulation. Fetal steroidogenesis ceases after sex differentiation is completed, and some fetal Leydig cells dedifferentiate to serve as stem cells for adult testicular cell types. Significant gaps are acknowledged: (1) Why are adult and fetal Leydig cells different? (2) What are bona fide progenitor and fetal Leydig cell markers? (3) Which signaling pathways and transcription factors regulate fetal Leydig cell steroidogenesis? It is critical to discover answers to these questions so that we can understand vulnerable targets in fetal Leydig cells and the mechanisms for androgen production that when disrupted, leads to variations in sex differentiation that range from subtle to complete sex reversal.


Asunto(s)
Andrógenos , Células Intersticiales del Testículo , Animales , Masculino , Humanos , Células Intersticiales del Testículo/metabolismo , Andrógenos/metabolismo , Células Endoteliales/metabolismo , Proteínas Hedgehog/metabolismo , Testículo/metabolismo , Testosterona , Hormona Luteinizante/metabolismo , Receptores de HL/metabolismo , Mamíferos
2.
Development ; 147(6)2020 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-32108023

RESUMEN

Members of the Iroquois B (IrxB) homeodomain cluster genes, specifically Irx3 and Irx5, are crucial for heart, limb and bone development. Recently, we reported their importance for oocyte and follicle survival within the developing ovary. Irx3 and Irx5 expression begins after sex determination in the ovary but remains absent in the fetal testis. Mutually antagonistic molecular signals ensure ovary versus testis differentiation with canonical Wnt/ß-catenin signals paramount for promoting the ovary pathway. Notably, few direct downstream targets have been identified. We report that Wnt/ß-catenin signaling directly stimulates Irx3 and Irx5 transcription in the developing ovary. Using in silico analysis of ATAC- and ChIP-Seq databases in conjunction with mouse gonad explant transfection assays, we identified TCF/LEF-binding sequences within two distal enhancers of the IrxB locus that promote ß-catenin-responsive ovary expression. Meanwhile, Irx3 and Irx5 transcription is suppressed within the developing testis by the presence of H3K27me3 on these same sites. Thus, we resolved sexually dimorphic regulation of Irx3 and Irx5 via epigenetic and ß-catenin transcriptional control where their ovarian presence promotes oocyte and follicle survival vital for future ovarian health.


Asunto(s)
Epigénesis Genética/fisiología , Gónadas/embriología , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Gónadas/metabolismo , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Ovario/embriología , Ovario/metabolismo , Caracteres Sexuales , Diferenciación Sexual/genética , Testículo/embriología , Testículo/metabolismo , Factores de Transcripción/metabolismo
3.
PLoS Genet ; 16(6): e1008810, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32497091

RESUMEN

Urogenital tract abnormalities are among the most common congenital defects in humans. Male urogenital development requires Hedgehog-GLI signaling and testicular hormones, but how these pathways interact is unclear. We found that Gli3XtJ mutant mice exhibit cryptorchidism and hypospadias due to local effects of GLI3 loss and systemic effects of testicular hormone deficiency. Fetal Leydig cells, the sole source of these hormones in developing testis, were reduced in numbers in Gli3XtJ testes, and their functional identity diminished over time. Androgen supplementation partially rescued testicular descent but not hypospadias in Gli3XtJ mutants, decoupling local effects of GLI3 loss from systemic effects of androgen insufficiency. Reintroduction of GLI3 activator (GLI3A) into Gli3XtJ testes restored expression of Hedgehog pathway and steroidogenic genes. Together, our results show a novel function for the activated form of GLI3 that translates Hedgehog signals to reinforce fetal Leydig cell identity and stimulate timely INSL3 and testosterone synthesis in the developing testis. In turn, exquisite timing and concentrations of testosterone are required to work alongside local GLI3 activity to control development of a functionally integrated male urogenital tract.


Asunto(s)
Criptorquidismo/genética , Regulación del Desarrollo de la Expresión Génica , Células Intersticiales del Testículo/patología , Proteínas del Tejido Nervioso/metabolismo , Diferenciación Sexual/genética , Proteína Gli3 con Dedos de Zinc/metabolismo , Animales , Criptorquidismo/patología , Modelos Animales de Enfermedad , Proteínas Hedgehog/metabolismo , Humanos , Insulina/metabolismo , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas/metabolismo , Transducción de Señal/genética , Testosterona/metabolismo , Proteína Gli3 con Dedos de Zinc/genética
4.
Biol Reprod ; 106(5): 1000-1010, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35138358

RESUMEN

Appropriate embryo-uterine interactions are essential for implantation. Besides oocyte abnormalities, implantation failure is a major contributor to early pregnancy loss. Previously, we demonstrated that two members of the Iroquois homeobox transcription factor family, IRX3 and IRX5, exhibited distinct and dynamic expression profiles in the developing ovary to promote oocyte and follicle survival. Elimination of each gene independently caused subfertility, but with different breeding pattern outcomes. Irx3 KO (Irx3LacZ/LacZ) females produced fewer pups throughout their reproductive lifespan which could only be partially explained by poor oocyte quality. Thus, we hypothesized that IRX3 is also expressed in the uterus where it acts to support pregnancy. To test this hypothesis, we harvested pregnant uteri from control and Irx3 KO females to evaluate IRX3 expression profiles and the integrity of embryo implantation sites. Our results indicate that IRX3 is expressed in the endometrial stromal cells at day 4 of pregnancy (D4) with peak expression at D5-D6, and then greatly diminishes by D7. Further, studies showed that while embryos were able to attach to the uterus, implantation sites in Irx3 KO pregnant mice exhibited impaired vascularization and abnormal expression of decidualization markers. Finally, we also observed an impaired response of the Irx3 KO uteri to an artificial deciduogenic stimulus, indicating a critical role of this factor in regulating the decidualization program. Together, these data established that IRX3 promotes female fertility via at least two different mechanisms: (1) promoting competent oocytes and (2) facilitating functional embryo-uterine interactions during implantation.


Asunto(s)
Implantación del Embrión , Proteínas de Homeodominio , Factores de Transcripción , Útero , Animales , Comunicación , Decidua/metabolismo , Implantación del Embrión/fisiología , Femenino , Uniones Comunicantes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Embarazo , Células del Estroma/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Útero/metabolismo
5.
PLoS Genet ; 14(8): e1007488, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30071018

RESUMEN

Women and other mammalian females are born with a finite supply of oocytes that determine their reproductive lifespan. During fetal development, individual oocytes are enclosed by a protective layer of granulosa cells to form primordial follicles that will grow, mature, and eventually release the oocyte for potential fertilization. Despite the knowledge that follicles are dysfunctional and will die without granulosa cell-oocyte interactions, the mechanisms by which these cells establish communication is unknown. We previously identified that two members of the Iroquois homeobox transcription factor gene family, Irx3 and Irx5, are expressed within developing ovaries but not testes. Deletion of both factors (Irx3-Irx5EGFP/Irx3-Irx5EGFP) disrupted granulosa cell-oocyte contact during early follicle development leading to oocyte death. Thus, we hypothesized that Irx3 and Irx5 are required to develop cell-cell communication networks to maintain follicle integrity and female fertility. A series of Irx3 and Irx5 mutant mouse models were generated to assess roles for each factor. While both Irx3 and Irx5 single mutant females were subfertile, their breeding outcomes and ovary histology indicated distinct causes. Careful analysis of Irx3- and Irx5-reporter mice linked the cause of this disparity to dynamic spatio-temporal changes in their expression patterns. Both factors marked the progenitor pre-granulosa cell population in fetal ovaries. At the critical phase of germline nest breakdown and primordial follicle formation however, Irx3 and Irx5 transitioned to oocyte- and granulosa cell-specific expression respectively. Further investigation into the cause of follicle death in Irx3-Irx5EGFP/Irx3-Irx5EGFP ovaries uncovered specific defects in both granulosa cells and oocytes. Granulosa cell defects included poor contributions to basement membrane deposition and mis-localization of gap junction proteins. Granulosa cells and oocytes both presented fewer cell projections resulting in compromised cell-cell communication. Altogether, we conclude that Irx3 and Irx5 first work together to define the pregranulosa cell population of germline nests. During primordial follicle formation, they transition to oocyte- and granulosa cell-specific expression patterns where they cooperate in neighboring cells to build the foundation for follicle integrity. This foundation is left as their legacy of the essential oocyte-granulosa cell communication network that ensures and ultimately optimizes the integrity of the ovarian reserve and therefore, the female reproductive lifespan.


Asunto(s)
Células de la Granulosa/fisiología , Proteínas de Homeodominio/fisiología , Factores de Transcripción/fisiología , Animales , Comunicación Celular , Conexinas/genética , Conexinas/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Germinativas , Proteínas de Homeodominio/genética , Ratones , Ratones Desnudos , Oocitos/fisiología , Factores de Transcripción/genética
6.
Biol Reprod ; 103(3): 620-629, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32507881

RESUMEN

Healthy development of ovarian follicles depends on appropriate interactions and function between oocytes and their surrounding granulosa cells. Previously, we showed that double knockout of Irx3 and Irx5 (Irx3/5 DKO) in mice resulted in abnormal follicle morphology and follicle death. Further, female mouse models of individual Irx3 or Irx5 knockouts were both subfertile but with distinct defects. Notably, the expression profile of each gene suggests independent roles for each; first, they are colocalized in pre-granulosa cells during development that then progresses to include oocyte expression during germline nest breakdown and primordial follicle formation. Thereafter, their expression patterns diverge between oocytes and granulosa cells coinciding with the formulation and maturation of intimate oocyte-granulosa cell interactions. The objective of this study was to investigate the contributions of Irx5 and somatic cell-specific expression of Irx3 during ovarian development. Our results show that Irx3 and Irx5 contribute to female fertility through different mechanisms and that Irx3 expression in somatic cells is important for oocyte quality and survival. Based on evaluation of a series of genetically modified mouse models, we conclude that IRX3 and IRX5 collaborate in the same cells and then in neighboring cells to foster a healthy and responsive follicle. Long after these two factors have extinguished, their legacy enables these intercellular connections to mature and respond to extracellular signals to promote follicle maturation and ovulation.


Asunto(s)
Células de la Granulosa/fisiología , Proteínas de Homeodominio/genética , Folículo Ovárico/crecimiento & desarrollo , Ovario/crecimiento & desarrollo , Factores de Transcripción/genética , Animales , Femenino , Fertilidad/genética , Infertilidad/genética , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Folículo Ovárico/citología , Ovario/citología , Embarazo , Diferenciación Sexual
7.
Vet Radiol Ultrasound ; 56(3): 307-16, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25572121

RESUMEN

Equine carpal sheath effusion has multiple etiologies. The purpose of this retrospective study was to describe the prevalence of distinct musculoskeletal lesions lameness in a sample of horses with a clinical diagnosis of carpal sheath effusion. A total of 121 horses met inclusion criteria. Seventy-four percent (89/121) of horses were lame at presentation; middle-aged (9-18 years, 80%) and older (> 18 years, 85%) horses were lame more frequently than young horses (< 9 years, 44%). Ninety-three percent (113/121) were diagnosed with osseous and/or soft tissue abnormalities. Of these 113 horses, 10 exhibited osseous abnormalities, whereas 111 were diagnosed with soft tissue lesions. Eighty-four percent (93/111) of the soft tissue injuries extended from the caudodistal antebrachium to the palmar metacarpus. The superficial digital flexor tendon (98/111; 88%) and accessory ligament of the superficial digital flexor tendon (64/111; 58%) were the most commonly injured structures, with both structures affected in 41 (41/111; 37%) horses. Injuries within the caudodistal antebrachium included the superficial digital flexor musculotendinous junction (66), the accessory ligament of the superficial digital flexor tendon (64), and deep digital flexor muscle (21), in isolation or in combination with other structures. Increased echogenicity in the medial superficial digital flexor musculotendinous junction was detected in 40 horses and was significantly associated with increasing age (middle-aged, 19/40; old, 18/40). Findings from this study indicated that age should be taken into consideration for horses presented with carpal sheath effusion and that adjacent structures within the caudodistal antebrachium should be included in evaluations.


Asunto(s)
Enfermedades de los Caballos/diagnóstico por imagen , Cojera Animal/diagnóstico por imagen , Animales , Estudios Transversales , Femenino , Marcha , Enfermedades de los Caballos/epidemiología , Caballos , Cojera Animal/epidemiología , Masculino , Prevalencia , Radiografía , Estudios Retrospectivos , Tendones/diagnóstico por imagen , Ultrasonografía , Estados Unidos/epidemiología
8.
Biol Reprod ; 91(4): 85, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25143354

RESUMEN

Despite the fact that fetal Leydig cells are recognized as the primary source of androgens in male embryos, the mechanisms by which steroidogenesis occurs within the developing testis remain unclear. A genetic approach was used to visualize and isolate fetal Leydig cells from remaining cells within developing mouse testes. Cyp11a1-Cre mice were bred to mT/mG dual reporter mice to target membrane-tagged enhanced green fluorescent protein (GFP) within steroidogenic cells, whereas other cells expressed membrane-tagged tandem-dimer tomato red. Fetal Leydig cell identity was validated using double-labeled immunohistochemistry against GFP and the steroidogenic enzyme 3beta-HSD, and cells were successfully isolated as indicated by qPCR results from sorted cell populations. Because fetal Leydig cells must collaborate with neighboring cells to synthesize testosterone, we hypothesized that the fetal Leydig cell microenvironment defined their capacity for androgen production. Microfluidic culture devices were used to measure androstenedione and testosterone production of fetal Leydig cells that were cultured in cell-cell contact within a mixed population, were isolated but remained in medium contact via compartmentalized co-culture with other testicular cells, or were isolated and cultured alone. Results showed that fetal Leydig cells maintained their identity and steroidogenic activity for 3-5 days in primary culture. Microenvironment dictated proficiency of testosterone production. As expected, fetal Leydig cells produced androstenedione but not testosterone when cultured in isolation. More testosterone accumulated in medium from mixed cultures than from compartmentalized co-cultures initially; however, co-cultures maintained testosterone synthesis for a longer time. These data suggest that a combination of cell-cell contact and soluble factors constitute the ideal microenvironment for fetal Leydig cell activity in primary culture.


Asunto(s)
Andrógenos/metabolismo , Embrión de Mamíferos/citología , Células Intersticiales del Testículo/metabolismo , Androstenos , Animales , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Medios de Cultivo , Regulación de la Expresión Génica , Masculino , Ratones , Técnicas Analíticas Microfluídicas , Comunicación Paracrina
9.
Endocrinology ; 165(5)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38597659

RESUMEN

We examined the effect of the puberty blocker, leuprolide acetate, on sex differences in juvenile rough-and-tumble play behavior and anxiety-like behavior in adolescent male and female rats. We also evaluated leuprolide treatment on gonadal and pituitary hormone levels and activity-regulated cytoskeleton-protein messenger RNA levels within the adolescent amygdala, a region important both for rough-and-tumble play and anxiety-like behavior. Our findings suggest that leuprolide treatment lowered anxiety-like behavior during adolescent development, suggesting that the maturation of gonadotropin-releasing hormone systems may be linked to increased anxiety. These data provide a potential new model to understand the emergence of increased anxiety triggered around puberty. Leuprolide also reduced masculinized levels of rough-and-tumble play behavior, lowered follicle-stimulating hormone, and produced a consistent pattern of reducing or halting sex differences of hormone levels, including testosterone, growth hormone, thyrotropin, and corticosterone levels. Therefore, leuprolide treatment not only pauses sexual development of peripheral tissues, but also reduces sex differences in hormones, brain, and behavior, allowing for better harmonization of these systems following gender-affirming hormone treatment. These data contribute to the intended use of puberty blockers in stopping sex differences from developing further with the potential benefit of lowering anxiety-like behavior.


Asunto(s)
Ansiedad , Conducta Animal , Leuprolida , Maduración Sexual , Animales , Leuprolida/farmacología , Masculino , Femenino , Ansiedad/tratamiento farmacológico , Ratas , Conducta Animal/efectos de los fármacos , Maduración Sexual/efectos de los fármacos , Caracteres Sexuales , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Corticosterona/sangre , Ratas Sprague-Dawley , Testosterona/sangre
10.
Biol Reprod ; 89(2): 34, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23843232

RESUMEN

MicroRNAs are important regulators of developmental gene expression, but their contribution to fetal gonad development is not well understood. We have identified the evolutionarily conserved gonadal microRNAs miR-202-5p and miR-202-3p as having a potential role in regulating mouse embryonic gonad differentiation. These microRNAs are expressed in a sexually dimorphic pattern as the primordial XY gonad differentiates into a testis, with strong expression in Sertoli cells. In vivo, ectopic expression of pri-miR-202 in XX gonads did not result in molecular changes to the ovarian determination pathway. Expression of the primary transcript of miR-202-5p/3p remained low in XY gonads in a conditional Sox9-null mouse model, suggesting that pri-miR-202 transcription is downstream of SOX9, a transcription factor that is both necessary and sufficient for male sex determination. We identified the pri-miR-202 promoter that is sufficient to drive expression in XY but not XX fetal gonads ex vivo. Mutation of SOX9 and SF1 binding sites reduced ex vivo transactivation of the pri-miR-202 promoter, demonstrating that pri-miR-202 may be a direct transcriptional target of SOX9/SF1 during testis differentiation. Our findings indicate that expression of the conserved gonad microRNA, miR-202-5p/3p, is downstream of the testis-determining factor SOX9, suggesting an early role in testis development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Organogénesis/genética , Factor de Transcripción SOX9/metabolismo , Testículo/embriología , Animales , Diferenciación Celular/genética , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Regiones Promotoras Genéticas , Factor de Transcripción SOX9/genética , Células de Sertoli/citología , Células de Sertoli/metabolismo , Diferenciación Sexual/genética , Testículo/citología , Testículo/metabolismo , Transcripción Genética
11.
Mol Reprod Dev ; 80(12): 960-76, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24105719

RESUMEN

The ovary functions to chaperone the most precious cargo for female individuals, the oocyte, thereby allowing the passage of genetic material to subsequent generations. Within the ovary, single oocytes are surrounded by a legion of granulosa cells inside each follicle. These two cell types depend upon one another to support follicle formation and oocyte survival. The infrastructure and events that work together to ultimately form these functional follicles within the ovary are unprecedented, given that the oocyte originates as a cell like all other neighboring cells within the embryo prior to gastrulation. This review discusses the journey of the germ cell in the context of the developing female mouse embryo, with a focus on specific signaling events and cell-cell interactions that escort the primordial germ cell as it is specified into the germ cell fate, migrates through the hindgut into the gonad, differentiates into an oocyte, and culminates upon formation of the primordial and then primary follicle.


Asunto(s)
Células de la Granulosa/citología , Oocitos/citología , Folículo Ovárico/citología , Folículo Ovárico/embriología , Animales , Proteínas Cdh1/metabolismo , Comunicación Celular , Diferenciación Celular , Movimiento Celular , Femenino , Ratones , Oocitos/metabolismo , Transducción de Señal
12.
Dev Biol ; 349(2): 417-26, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20969841

RESUMEN

Over the first 4 days of their life, primordial germ cells invade the endoderm, migrate into and through the developing hindgut, and traverse to the genital ridge where they cluster and ultimately inhabit the nascent gonad. Specific signal-receptor combinations between primordial germ cells and their immediate environment establish successful migration and colonization. Here we demonstrate that disruption of a cluster of six genes on murine chromosome 8, as exemplified by the Fused Toes (Ft) mutant mouse model, results in severely decreased numbers of primordial germ cells within the early gonad. Primordial germ cell migration appeared normal within Ft mutant embryos; however, germ cell counts progressively decreased during this time. Although no difference in apoptosis was detected, we report a critical decrease in primordial germ cell proliferation by E12.5. The six genes within the Ft locus include the IrxB cluster (Irx3, -5, -6), Fts, Ftm, and Fto, of which only Ftm, Fto, and Fts are expressed in primordial germ cells of the early gonad. From these studies, we have discovered that the Ft locus on mouse chromosome 8 is associated with cell cycle deficits within the primordial germ cell population that initiates just before translocation into the genital ridge.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular , Células Germinativas/fisiología , Gónadas/embriología , Proteínas/genética , Animales , Proteínas Reguladoras de la Apoptosis , Recuento de Células , Cartilla de ADN/genética , Femenino , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Masculino , Ratones , Mutación/genética , Reacción en Cadena de la Polimerasa
13.
Biol Reprod ; 84(5): 1024-32, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21293032

RESUMEN

Ovarian development absolutely depends on communication between somatic and germ cell components. In contrast, it is not until after birth that interactions between somatic and germ cells play an important role in testicular maturation and spermatogenesis. Previously, we discovered that Irx3 expression was localized specifically to female gonads during embryonic development; therefore, we sought to determine the function of this genetic locus in developing gonads of both sexes. The fused toes (Ft) mutant mouse is missing 1.6 Mb of chromosome 8, which includes the entire IrxB cluster (Irx3, Irx5, Irx6), Ftm, Fts, and Fto genes. Homozygote Ft mutant embryos die around embryonic day 13.5 (E13.5); therefore, to assess later development, we harvested gonads at E11.5 and transplanted them into nude mouse hosts. Our results show defects in somatic and germ cell maturation in developing gonads of both sexes. Testis development was normal initially; however, by 3-wk posttransplantation, expression of Sertoli and peritubular myoid cell markers were decreased. In many cases, gonocytes failed to migrate to structurally impaired basement membranes of seminiferous cords. Developmental abnormalities of the ovary appeared earlier and were more severe. Over time, the Ft mutant ovary formed very few primordial or primary follicles, which contained oocytes that failed to grow and were surrounded by scarce granulosa cells that expressed low levels of FOXL2. By 3 wk after transplantation, it was difficult to identify ovarian tissue in Ft mutant ovary transplants. In summary, we conclude that the Ft locus contains genes essential for somatic-germ cell interactions, without which the germ cell niche fails to mature in both sexes.


Asunto(s)
Anomalías Múltiples/veterinaria , Deleción Cromosómica , Cromosomas de los Mamíferos , Gónadas/embriología , Proteínas de Homeodominio/genética , Enfermedades de los Roedores/genética , Factores de Transcripción/genética , Anomalías Múltiples/genética , Animales , Antígenos de Diferenciación/metabolismo , Comunicación Celular , Diferenciación Celular , Movimiento Celular , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Gónadas/citología , Gónadas/patología , Gónadas/trasplante , Células de la Granulosa/metabolismo , Células de la Granulosa/patología , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Mutantes , Ratones Desnudos , Células de Sertoli/metabolismo , Células de Sertoli/patología , Caracteres Sexuales , Dedos del Pie/anomalías , Dedos del Pie/embriología , Factores de Transcripción/metabolismo
14.
Biol Reprod ; 84(3): 422-34, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20962249

RESUMEN

Targets of steroidogenic factor 1 (SF1; also known as NR5A1 and AD4BP) have been identified within cells at every level of the hypothalamic-pituitary-gonadal and -adrenal axes, revealing SF1 to be a master regulator of major endocrine systems. Mouse embryos express SF1 in the genital ridge until Embryonic Day 13.5 (E13.5). Thereafter, expression persists in the male and is substantially lower in the female gonad until birth. We hypothesize that the sexually dimorphic expression of Sf1 during gonadogenesis is mediated by sex-specific regulation of its promoter. To investigate dimorphic regulation within the fetal gonad, we developed an experimental strategy using transient transfection of E13.5 gonad explant cultures and evaluated various Sf1 promoter constructs for sexually dimorphic DNA elements. The proximal Sf1 promoter correctly targeted reporter activity to SF1-expressing cells in both XY and XX gonads. Stepwise deletion of sequences from the Sf1 promoter revealed two regions that affected regulation within female gonads. Mutation of both sequences together did not cause further disruption of reporter activity, suggesting the two sites might work in concert to promote activity in female somatic cells. Results from gel mobility shift assays and fetal gonad-chromatin immunoprecipitation showed that TCFAP2 binds to one of the two female-specific sites within the proximal promoter of Sf1. Together, we show that transient transfection experiments performed within developing testes and ovaries are a powerful tool to uncover elements within the Sf1 promoter that contribute to sex-specific expression.


Asunto(s)
Ovario/embriología , Ovario/metabolismo , Regiones Promotoras Genéticas/fisiología , Factor Esteroidogénico 1/genética , Animales , Secuencia de Bases , Células Cultivadas , Electroporación , Femenino , Gónadas/embriología , Gónadas/metabolismo , Masculino , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Especificidad de Órganos/genética , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Factor Esteroidogénico 1/metabolismo
15.
Endocrinology ; 162(7)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33784378

RESUMEN

Two specialized functions of cholesterol during fetal development include serving as a precursor to androgen synthesis and supporting hedgehog (HH) signaling activity. Androgens are produced by the testes to facilitate masculinization of the fetus. Recent evidence shows that intricate interactions between the HH and androgen signaling pathways are required for optimal male sex differentiation and defects of either can cause birth anomalies indicative of 46,XY male variations of sex development (VSD). Further, perturbations in cholesterol synthesis can cause developmental defects, including VSD, that phenocopy those caused by disrupted androgen or HH signaling, highlighting the functional role of cholesterol in promoting male sex differentiation. In this review, we focus on the role of cholesterol in systemic androgen and local HH signaling events during fetal masculinization and their collective contributions to pediatric VSD.


Asunto(s)
Andrógenos/biosíntesis , Colesterol/fisiología , Proteínas Hedgehog/metabolismo , Diferenciación Sexual/fisiología , Transducción de Señal/fisiología , Animales , Colesterol/biosíntesis , Trastornos del Desarrollo Sexual , Desarrollo Fetal/fisiología , Feto/metabolismo , Humanos , Células Intersticiales del Testículo/fisiología , Masculino , Testículo/embriología , Testículo/metabolismo
16.
Mol Cell Endocrinol ; 531: 111265, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33864885

RESUMEN

STARD1 stimulates cholesterol transfer to mitochondrial CYP11A1 for conversion to pregnenolone. A cholesterol-binding START domain is guided by an N-terminal domain in a cell selective manner. Fetal and adult Leydig cells (FLC, ALC) show distinct Stard1 regulation. sm- FISH microscopy, which resolves individual molecules of Stard1 mRNA, shows uniformly high basal expression in each FLC. In ALC, in vivo, and cultured MA-10 cells, basal Stard1 expression is minimal. PKA activates loci asynchronously, with delayed splicing/export of 3.5 kb mRNA to mitochondria. After 60 min, ALC transition to an integrated mRNA delivery to mitochondria that is seen in FLC. Sertoli cells cooperate in Stard1 stimulation in FLC by delivering DHH to the primary cilium. There PTCH, SMO and cholesterol cooperate to release GLI3 to activate the Stard1 locus, probably by directing histone changes. ALC lack cilia. PKA then primes locus activation. FLC and ALC share similar SIK/CRTC/CREB regulation characterized for adrenal cells.


Asunto(s)
Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Células Intersticiales del Testículo/fisiología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Empalme Alternativo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Masculino , Fosfoproteínas/química , Dominios Proteicos , Transducción de Señal , Imagen Individual de Molécula
17.
Dev Biol ; 329(1): 96-103, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19268447

RESUMEN

Proper cell fate determination in mammalian gonads is critical for the establishment of sexual identity. The Hedgehog (Hh) pathway has been implicated in cell fate decision for various organs, including gonads. Desert Hedgehog (Dhh), one of the three mammalian Hh genes, has been implicated with other genes in the establishment of mouse fetal Leydig cells. To investigate whether Hh alone is sufficient to induce fetal Leydig cell differentiation, we ectopically activated the Hh pathway in Steroidogenic factor 1 (SF1)-positive somatic cell precursors of fetal ovaries. Hh activation transformed SF1-positive somatic ovarian cells into functional fetal Leydig cells. These ectopic fetal Leydig cells produced androgens and insulin-like growth factor 3 (INLS3) that cause virilization of female embryos and ovarian descent. However, the female reproductive system remained intact, indicating a typical example of female pseudohermaphroditism. The appearance of fetal Leydig cells was a direct consequence of Hh activation as evident by the absence of other testicular components in the affected ovary. This study provides not only insights into mechanisms of cell lineage specification in gonads, but also a model to understand defects in sexual differentiation.


Asunto(s)
Trastornos del Desarrollo Sexual/metabolismo , Feto , Proteínas Hedgehog/metabolismo , Células Intersticiales del Testículo/metabolismo , Ovario/metabolismo , Animales , Diferenciación Celular/genética , Trastornos del Desarrollo Sexual/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Feto/citología , Feto/metabolismo , Proteínas Hedgehog/genética , Inmunohistoquímica , Hibridación in Situ , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/fisiología , Masculino , Ratones , Ratones Transgénicos , Diferenciación Sexual , Transducción de Señal/genética
18.
Front Endocrinol (Lausanne) ; 11: 559674, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193082

RESUMEN

STARD1 moves cholesterol (CHOL) from the outer mitochondrial membrane (OMM) to the inner membrane (IMM) in steroidogenic cells. This activity is integrated into CHOL trafficking and synthesis homeostasis, involving uptake through SR-B1 and LDL receptors and distribution through endosomes, ER, and lipid droplets. In adrenal cells, STARD1 is imported into the mitochondrial matrix accompanied by delivery of several hundred CHOL molecules. This transfer limits CYP11A1-mediated generation of pregnenolone. CHOL transfer is coupled to translation of STARD1 mRNA at the OMM. In testis cells, slower CHOL trafficking seems to be limiting. STARD1 also functions in a slower process through ER OMM contacts. The START domain of STARD1 is utilized by a family of genes, which includes additional STARD (forms 3-6) and GRAMD1B proteins that transfer CHOL. STARD forms 2 and 7 deliver phosphatidylcholine. STARD1 and STARD7 target their respective activities to mitochondria, via N-terminal domains (NTD) of over 50 amino acids. The NTD is not essential for steroidogenesis but exerts tissue-selective enhancement (testis>>adrenal). Three conserved sites for cleavage by the mitochondrial processing protease (MPP) generate three forms, each potentially with specific functions, as demonstrated in STARD7. STARD1 is expressed in macrophage and cardiac repair fibroblasts. Additional functions include CHOL metabolism by CYP27A1 that directs activation of LXR and CHOL export processes. STARD1 generates 3.5- and 1.6-kb mRNA from alternative polyadenylation. The 3.5-kb form exclusively binds the PKA-induced regulator, TIS11b, which binds at conserved sites in the extended 3'UTR to control mRNA translation and turnover. STARD1 expression also exhibits a novel, slow splicing that delayed splicing delivery of mRNA to mitochondria. Stimulation of transcription by PKA is directed by suppression of SIK forms that activate a CRTC/CREB/CBP promoter complex. This process is critical to pulsatile hormonal activation in vivo. sm-FISH RNA imaging shows a flow of single STARD1 mRNA particles from asymmetric accumulations of primary transcripts at gene loci to 1:1 complex of 3.5-kb mRNA with peri-nuclear mitochondria. Adrenal cells are similar but distinguished from testis cells by appreciable basal expression prior to hormonal activation. This difference is conserved in culture and in vivo.


Asunto(s)
HDL-Colesterol/metabolismo , Mitocondrias/metabolismo , Imagen Molecular/métodos , Fosfoproteínas/metabolismo , Empalme del ARN/fisiología , ARN Mensajero/biosíntesis , Animales , Colesterol/genética , Colesterol/metabolismo , HDL-Colesterol/genética , Expresión Génica , Humanos , Mitocondrias/genética , Membranas Mitocondriales/metabolismo , Fosfoproteínas/genética , ARN Mensajero/genética
19.
Lab Chip ; 20(1): 107-119, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31712791

RESUMEN

Open microfluidic cell culture systems are powerful tools for interrogating biological mechanisms. We have previously presented a microscale cell culture system, based on spontaneous capillary flow of biocompatible hydrogels, that is integrated into a standard cell culture well plate, with flexible cell compartment geometries and easy pipet access. Here, we present two new injection molded open microfluidic devices that also easily insert into standard cell culture well plates and standard culture workflows, allowing seamless adoption by biomedical researchers. These platforms allow culture and study of soluble factor communication among multiple cell types, and the microscale dimensions are well-suited for rare primary cells. Unique advances include optimized evaporation control within the well, manufacture with reproducible and cost-effective rapid injection molding, and compatibility with sample preparation workflows for high resolution microscopy (following well-established coverslip mounting procedures). In this work, we present several use cases that highlight the usability and widespread utility of our platform including culture of limited primary testis cells from surgical patients, microscopy readouts including immunocytochemistry and single molecule fluorescence in situ hybridization (smFISH), and coculture to study interactions between adipocytes and prostate cancer cells.


Asunto(s)
Dispositivos Laboratorio en un Chip , Testículo/citología , Supervivencia Celular , Células Cultivadas , Técnicas de Cocultivo , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino
20.
Endocr Rev ; 25(4): 521-42, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15294880

RESUMEN

Normal reproductive function in mammals requires precise control of LH synthesis and secretion by gonadotropes of the anterior pituitary. Synthesis of LH requires expression of two genes [alpha-glycoprotein subunit (alphaGSU) and LHbeta] located on different chromosomes. Hormones from the hypothalamus and gonads modulate transcription of both genes as well as secretion of the biologically active LH heterodimer. In males and females, the transcriptional tone of the genes encoding alphaGSU and LHbeta reflects dynamic integration of a positive signal provided by GnRH from hypothalamic neurons and negative signals emanating from gonadal steroids. Although alphaGSU and LHbeta genes respond transcriptionally in the same manner to changes in hormonal input, different combinations of regulatory elements orchestrate their response. These hormone-responsive regulatory elements are also integral members of much larger combinatorial codes responsible for targeting expression of alphaGSU and LHbeta genes to gonadotropes. In this review, we will profile the genomic landscape of the promoter-regulatory region of both genes, depicting elements and factors that contribute to gonadotrope-specific expression and hormonal regulation. Within this context, we will highlight the different combinatorial codes that control transcriptional responses, particularly those that mediate the opposing effects of GnRH and one of the sex steroids, androgens. We will use this framework to suggest that GnRH and androgens attain the same transcriptional endpoint through combinatorial codes unique to alphaGSU and LHbeta. This parallelism permits the dynamic and coordinate regulation of two genes that encode a single hormone.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Código Genético , Hormona Luteinizante/genética , Regiones Promotoras Genéticas/fisiología , Secuencias Reguladoras de Ácidos Nucleicos/fisiología , Animales , Femenino , Humanos , Hormona Luteinizante/metabolismo , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA