Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biol Reprod ; 110(6): 1055-1064, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38315794

RESUMEN

Reproductive processes are dynamic and involve extensive morphological remodeling and cell-cell interactions. Live imaging of organs enhances our understanding of how biological processes occur in real time. Slice culture is a type of organ culture where thick slices are collected from an organ and cultured for several days. Slice culture is a useful and easy-to-implement technique for live imaging of reproductive events at cellular resolution. Here we describe a pipeline of live imaging on slice culture to visualize the process of urethra closure in mouse embryonic penis as a proof of principle. In combination with genetic reporter mice, nuclear stains, and exposure experiments, we demonstrate the feasibility of slice culture on a reproductive organ. We also provide a step-by-step protocol and troubleshooting guide to facilitate the adoption of slice culture with live imaging in other reproductive organs. Lastly, we discuss potential utilities and experiments that could be implemented with slice culture in reproductive sciences.


Asunto(s)
Técnicas de Cultivo de Órganos , Animales , Técnicas de Cultivo de Órganos/métodos , Ratones , Masculino , Reproducción/fisiología , Uretra , Pene , Femenino
2.
bioRxiv ; 2023 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-37986842

RESUMEN

Hypospadias, or incomplete closure of the urethra along the penis, is the second most common birth defect in the United States. We discovered a population of extra-genital mesenchymal cells that are essential for proper penile urethra closure in mouse embryos. This extra-genital population first appeared in the mesenchyme posterior to the hindlimb of the fetus after the onset of penis formation. These extra-genital cells, which transiently express a lineage marker Nr5a1, migrated centrally and colonized the penis bilateral to the urethra epithelium. Removal of the Nr5a1+ extra-genital cells, using a cell-type specific ablation model, resulted in severe hypospadias. The absence of extra-genital cells had the most significant impacts on another mesenchymal cells, the peri-urethra that were immediately adjacent to the Nr5a1+ extra-genital cells. Single cell mRNA sequencing revealed that the extra-genital cells extensively interact with the peri-urethra, particularly through Neuregulin 1, an epidermal Growth Factor (EGF) ligand. Disruption of Neuregulin 1 signaling in the ex-vivo slice culture system led to failure of urethra closure, recapitulating the phenotypes of extra-genital cell ablation. These results demonstrate that the Nr5a1+ extra-genital mesenchymal cells from outside of the fetal penis are indispensable for urethra closure through their interaction with the peri-urethra mesenchymal cells. This discovery provides a new entry point to understand the biology of penis formation and potential causes of hypospadias in humans.

3.
Biol Reprod ; 108(6): 866-870, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-36951956

RESUMEN

Sertoli cells, first identified in the adult testis by Enrico Sertoli in the mid-nineteenth century, are known for their role in fostering male germ cell differentiation and production of mature sperm. It was not until the late twentieth century with the discovery of the testis-determining gene SRY that Sertoli cells' new function as the master regulator of testis formation and maleness was unveiled. Fetal Sertoli cells facilitate the establishment of seminiferous cords, induce appearance of androgen-producing Leydig cells, and cause regression of the female reproductive tracts. Originally thought be a terminally differentiated cell type, adult Sertoli cells, at least in the mouse, retain their plasticity and ability to transdifferentiate into the ovarian counterpart, granulosa cells. In this review, we capture the many phases of Sertoli cell differentiation from their fate specification in fetal life to fate maintenance in adulthood. We also introduce the discovery of a new phase of fetal Sertoli cell differentiation via autocrine/paracrine factors with the freemartin characteristics. There remains much to learn about this intriguing cell type that lay the foundation for the maleness.


Asunto(s)
Freemartinismo , Testículo , Bovinos , Masculino , Femenino , Animales , Ratones , Testículo/metabolismo , Freemartinismo/metabolismo , Semen , Células de Sertoli/metabolismo , Células Intersticiales del Testículo/metabolismo
4.
PNAS Nexus ; 1(4): pgac182, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36204418

RESUMEN

The female reproductive tract develops from its embryonic precursor, the Müllerian duct. In close proximity to the Müllerian duct lies the precursor for the male reproductive tract, the Wolffian duct, which is eliminated in the female embryo during sexual differentiation. We discovered that a component of the Wolffian duct, its mesenchyme, is not eliminated after sexual differentiation. Instead, the Wolffian duct mesenchyme underwent changes in transcriptome and chromatin accessibility from male tract to female tract identity, and became a unique mesenchymal population in the female reproductive tract with localization and transcriptome distinct from the mesenchyme derived from the Müllerian duct. Partial ablation of the Wolffian duct mesenchyme stunted the growth of the fetal female reproductive tract in ex vivo organ culture. These findings reveal a new fetal origin of mesenchymal tissues for female reproductive tract formation and reshape our understanding of sexual differentiation of reproductive tracts.

5.
Nat Commun ; 13(1): 4130, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35840551

RESUMEN

Fate determination and maintenance of fetal testes in most mammals occur cell autonomously as a result of the action of key transcription factors in Sertoli cells. However, the cases of freemartin, where an XX twin develops testis structures under the influence of an XY twin, imply that hormonal factor(s) from the XY embryo contribute to sex reversal of the XX twin. Here we show that in mouse XY embryos, Sertoli cell-derived anti-Mullerian hormone (AMH) and activin B together maintain Sertoli cell identity. Sertoli cells in the gonadal poles of XY embryos lacking both AMH and activin B transdifferentiate into their female counterpart granulosa cells, leading to ovotestis formation. The ovotestes remain to adulthood and produce both sperm and oocytes, although there are few of the former and the latter fail to mature. Finally, the ability of XY mice to masculinize ovaries is lost in the absence of these two factors. These results provide insight into fate maintenance of fetal testes through the action of putative freemartin factors.


Asunto(s)
Activinas , Hormona Antimülleriana , Diferenciación Celular , Testículo , Activinas/metabolismo , Activinas/farmacología , Animales , Hormona Antimülleriana/metabolismo , Hormona Antimülleriana/farmacología , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/fisiología , Diferenciación Celular/fisiología , Femenino , Masculino , Mamíferos , Ratones , Comunicación Paracrina/fisiología , Semen , Células de Sertoli , Testículo/metabolismo
6.
FASEB J ; 35(9): e21876, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34449112

RESUMEN

Compared with the well-described XY sex determination system in mammals, the avian ZW sex determination system is poorly understood. Knockdown and overexpression studies identified doublesex and mab-3-related transcription factor 1 (DMRT1) as the testis-determining gene in chicken. However, the detailed effects of DMRT1 gene disruption from embryonic to adult development are not clear. Herein, we have generated DMRT1-disrupted chickens using the clustered regularly interspaced short palindromic repeats-associated protein 9 system, followed by an analysis of physiological, hormonal, and molecular changes in the genome-modified chickens. In the early stages of male chicken development, disruption of DMRT1 induced gonad feminization with extensive physiological and molecular changes; however, functional feminine reproductivity could not be implemented with disturbed hormone synthesis. Subsequent RNA-sequencing analysis of the DMRT1-disrupted chicken gonads revealed gene networks, including several novel genes linearly and non-linearly associated with DMRT1, which are involved in gonad feminization. By comparing the gonads of wild type with the genome-modified chickens, a set of genes were identified that is involved in the ZW sex determination system independent of DMRT1. Our results extend beyond the Z-dosage hypothesis to provide further information about the avian ZW sex determination system and epigenetic effects of gonad feminization.


Asunto(s)
Pollos/genética , Feminización/genética , Gónadas/fisiología , Factores de Transcripción/genética , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Masculino , Ovario/fisiología , Cromosomas Sexuales , Testículo/fisiología
7.
FASEB J ; 35(8): e21770, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34288113

RESUMEN

Steroid hormones regulate various aspects of physiology, from reproductive functions to metabolic homeostasis. Steroidogenic factor-1 (NR5A1) plays a central role in the development of steroidogenic tissues and their ability to produce steroid hormones. Inactivation of Nr5a1 in the mouse results in a complete gonadal and adrenal agenesis, absence of gonadotropes in the pituitary and impaired development of ventromedial hypothalamus, which controls glucose and energy metabolism. In this study, we set out to examine the consequences of NR5A1 overexpression (NR5A1+) in the NR5A1-positive cell populations in female mice. Ovaries of NR5A1+ females presented defects such as multi-oocyte follicles and an accumulation of corpora lutea. These females were hyperandrogenic, had irregular estrous cycles with persistent metestrus and became prematurely infertile. Furthermore, the decline in fertility coincided with weight gain, increased adiposity, hypertriglyceridemia, hyperinsulinemia, and impaired glucose tolerance, indicating defects in metabolic functions. In summary, excess NR5A1 expression causes hyperandrogenism, disruption of ovarian functions, premature infertility, and disorders of metabolic homeostasis. This NR5A1 overexpression mouse provides a novel model for studying not only the molecular actions of NR5A1, but also the crosstalk between endocrine, reproductive, and metabolic systems.


Asunto(s)
Fertilidad , Infertilidad/fisiopatología , Obesidad/fisiopatología , Ovario/fisiopatología , Factor Esteroidogénico 1/fisiología , Animales , Femenino , Homeostasis , Ratones , Ratones Endogámicos C57BL , Fenotipo
8.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34155146

RESUMEN

Birth defects of the external genitalia are among the most common in the world. Proper formation of the external genitalia requires a highly orchestrated process that involves special cell populations and sexually dimorphic hormone signaling. It is clear what the end result of the sexually dimorphic development is (a penis in the male versus clitoris in the female); however, the cell populations involved in the process remain poorly defined. Here, we used single-cell messenger RNA sequencing in mouse embryos to uncover the dynamic changes in cell populations in the external genitalia during the critical morphogenetic window. We found that overall, male and female external genitalia are largely composed of the same core cellular components. At the bipotential stage of development (embryonic day or E14.5), few differences in cell populational composition exist between male and female. Although similar in cell population composition, genetic differences in key sexual differentiation developmental pathways arise between males and females by the early (E16.5) and late (E18.5) differentiation stages. These differences include discrete cell populations with distinct responsiveness to androgen and estrogen. By late sexual differentiation (E18.5), unique cell populations in both male and female genitalia become apparent and are enriched with androgen- and estrogen-responsive genes, respectively. These data provide insights into the morphogenesis of the external genitalia that could be used to understand diseases associated with defects in the external genitalia.


Asunto(s)
Genitales/citología , Genitales/embriología , Caracteres Sexuales , Análisis de la Célula Individual , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Hormonas/metabolismo , Masculino , Mesodermo/citología , Mesodermo/embriología , Ratones Endogámicos C57BL , Modelos Biológicos
9.
Reprod Toxicol ; 95: 59-65, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32416200

RESUMEN

The flame retardant and plasticizer, tetrabromobisphenol-A (TBBPA) has rapidly become a common component in the manufacture of circuit boards and plastics worldwide. It is also an analog of bisphenol A (BPA), an endocrine disrupting chemical identified by the Endocrine Society. As such, TBBPA needs to be investigated for similar potential human health risks. Using rats as a model, we exposed pregnant dams and their progeny to 0, 0.1, 25, or 250 mg TBBPA/kg of body weight until the offspring reached adulthood and assessed the first generation of males for any reproductive tract abnormalities. We found no differences in the morphology of testes, sperm, prostates, or secondary sex organs from post-natal day 21 through one-year of age. A delay in the time to preputial separation was found with the 250 mg/kg treatment. Also, minor differences of sperm count at one-year old with the 25 mg/kg treatment and expression levels of two steroidogenic pathway enzymes at either post-natal day 90 or one-year old in the 250 mg/kg treatment group were detected, but spermatogenesis was not disrupted. While these results may lead to the supposition that TBBPA is less harmful than its parent compound BPA, more studies need to be conducted to assess long-term exposure effects.


Asunto(s)
Retardadores de Llama/toxicidad , Bifenilos Polibrominados/toxicidad , Animales , Femenino , Masculino , Intercambio Materno-Fetal , Embarazo , Próstata/efectos de los fármacos , Ratas Wistar , Salud Reproductiva , Recuento de Espermatozoides , Espermatozoides/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/metabolismo
10.
Biol Reprod ; 101(3): 602-616, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31058957

RESUMEN

Alfred Jost's work in the 1940s laid the foundation of the current paradigm of sexual differentiation of reproductive tracts, which contends that testicular hormones drive the male patterning of reproductive tract system whereas the female phenotype arises by default. Once established, the sex-specific reproductive tracts undergo morphogenesis, giving rise to anatomically and functionally distinct tubular organs along the rostral-caudal axis. Impairment of sexual differentiation of reproductive tracts by genetic alteration and environmental exposure are the main causes of disorders of sex development, and infertility at adulthood. This review covers past and present work on sexual differentiation and morphogenesis of reproductive tracts, associated human disorders, and emerging technologies that have made impacts or could radically expand our knowledge in this field.


Asunto(s)
Investigación Biomédica , Genitales/embriología , Morfogénesis/fisiología , Diferenciación Sexual/fisiología , Adulto , Animales , Investigación Biomédica/historia , Investigación Biomédica/métodos , Investigación Biomédica/tendencias , Femenino , Regulación del Desarrollo de la Expresión Génica , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Invenciones , Masculino , Reproducción/genética , Sistema Urogenital/embriología
11.
Endocr Rev ; 39(5): 739-759, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29771299

RESUMEN

The reproductive endocrine systems are vastly different between males and females. This sexual dimorphism of the endocrine milieu originates from sex-specific differentiation of the somatic cells in the gonads during fetal life. Most gonadal somatic cells arise from the adrenogonadal primordium. After separation of the adrenal and gonadal primordia, the gonadal somatic cells initiate sex-specific differentiation during gonadal sex determination with the specification of the supporting cell lineages: Sertoli cells in the testis vs granulosa cells in the ovary. The supporting cell lineages then facilitate the differentiation of the steroidogenic cell lineages, Leydig cells in the testis and theca cells in the ovary. Proper differentiation of these cell types defines the somatic cell environment that is essential for germ cell development, hormone production, and establishment of the reproductive tracts. Impairment of lineage specification and function of gonadal somatic cells can lead to disorders of sexual development (DSDs) in humans. Human DSDs and processes for gonadal development have been successfully modeled using genetically modified mouse models. In this review, we focus on the fate decision processes from the initial stage of formation of the adrenogonadal primordium in the embryo to the maintenance of the somatic cell identities in the gonads when they become fully differentiated in adulthood.


Asunto(s)
Linaje de la Célula/fisiología , Células de la Granulosa/citología , Ovario/citología , Células de Sertoli/citología , Testículo/citología , Animales , Diferenciación Celular/fisiología , Desarrollo Embrionario/fisiología , Femenino , Humanos , Masculino
12.
Biol Reprod ; 97(3): 365-377, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-29088396

RESUMEN

Despite its importance to reproduction, certain mechanisms of early ovarian development remain a mystery. To improve our understanding, we constructed the first cell-based computational model of ovarian development in mice that is divided into two phases: Phase I spans embryonic day 5.5 (E5.5) to E12.5; and Phase II spans E12.5 to postnatal day 2. We used the model to investigate four mechanisms: in Phase I, (i) whether primordial germ cells (PGCs) undergo mitosis during migration; and (ii) if the mechanism for secretion of KIT ligand from the hindgut resembles inductive cell-cell signaling or is secreted in a static manner; and in Phase II, (iii) that changes in cellular adhesion produce germ cell nest breakdown; and (iv) whether localization of primordial follicles in the cortex of the ovary is due to proliferation of granulosa cells. We found that the combination of the first three hypotheses produced results that aligned with experimental images and PGC abundance data. Results from the fourth hypothesis did not match experimental images, which suggests that more detailed processes are involved in follicle localization. Phase I and Phase II of the model reproduce experimentally observed cell counts and morphology well. A sensitivity analysis identified contact energies, mitotic rates, KIT chemotaxis strength, and diffusion rate in Phase I and oocyte death rate in Phase II as parameters with the greatest impact on model predictions. The results demonstrate that the computational model can be used to understand unknown mechanisms, generate new hypotheses, and serve as an educational tool.


Asunto(s)
Biología Computacional , Simulación por Computador , Ovario/crecimiento & desarrollo , Animales , Adhesión Celular , Movimiento Celular , Desarrollo Embrionario/fisiología , Femenino , Células Germinativas , Células de la Granulosa/fisiología , Ratones , Mitosis , Método de Montecarlo , Ovario/embriología , Embarazo , Diferenciación Sexual , Transducción de Señal/genética , Transducción de Señal/fisiología , Programas Informáticos , Factor de Células Madre
13.
Toxicol Sci ; 157(1): 8-19, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28123099

RESUMEN

Intrauterine exposure to phthalates is known to cause disorders of male reproductive function including androgen insufficiency, decreased fertility, and germ cell defects in rodents. In this study, we set out to investigate the effects of intrauterine exposure to di-(2-ethylhexyl)-phthalate (DEHP) on fetal development of the B6:129S4 mouse strain. Time-mated pregnant C57BL/6 dams were exposed to 0, 5, 250, or 500 mg/kg DEHP with corn oil as the vehicle via oral gavage from embryonic days (E)7 to 16. Survival and gross morphology of the pups were analyzed one day after the last treatment. Anogenital distance (AGD) and testicular cell functions were examined in male embryos to confirm the known effects of phthalate exposure. DEHP exposure significantly reduced the survival rate of fetuses in the 250 and 500 mg/kg dosage groups compared with the control and 5 mg/kg groups. Exposure to 250 and 500 mg/kg DEHP was teratogenic and induced exencephaly and limb malformations such as polydactyly in the B6:126S4 embryos. No gross malformations were observed in control or 5 mg/kg DEHP groups. In male embryos, exposure to both 5 and 250 mg/kg DEHP in utero was sufficient to induce the formation of multinucleated germ cells in the testes and widespread changes in mRNA expression of germ cell, interstitium and Sertoli cell-associated genes. These findings reveal that intrauterine DEHP exposure has a strong teratogenic, and lethal impact on the fetuses of B6:129S4 mouse strain.


Asunto(s)
Dietilhexil Ftalato/toxicidad , Desarrollo Fetal/efectos de los fármacos , Teratógenos/toxicidad , Anomalías Inducidas por Medicamentos , Animales , Relación Dosis-Respuesta a Droga , Femenino , Muerte Fetal , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Testículo/citología , Testículo/efectos de los fármacos , Testículo/embriología
15.
Environ Health Perspect ; 124(3): 336-43, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26295903

RESUMEN

BACKGROUND: Mice exposed to high levels of arsenic in utero have increased susceptibility to tumors such as hepatic and pulmonary carcinomas when they reach adulthood. However, the effects of in utero arsenic exposure on general physiological functions such as reproduction and metabolism remain unclear. OBJECTIVES: We evaluated the effects of in utero exposure to inorganic arsenic at the U.S. Environmental Protection Agency (EPA) drinking water standard (10 ppb) and at tumor-inducing levels (42.5 ppm) on reproductive end points and metabolic parameters when the exposed females reached adulthood. METHODS: Pregnant CD-1 mice were exposed to sodium arsenite [none (control), 10 ppb, or 42.5 ppm] in drinking water from gestational day 10 to birth, the window of organ formation. At birth, exposed offspring were fostered to unexposed dams. We examined reproductive end points (age at vaginal opening, reproductive hormone levels, estrous cyclicity, and fertility) and metabolic parameters (body weight changes, hormone levels, body fat content, and glucose tolerance) in the exposed females when they reached adulthood. RESULTS: Arsenic-exposed females (10 ppb and 42.5 ppm) exhibited early onset of vaginal opening. Fertility was not affected when females were exposed to the 10-ppb dose. However, the number of litters per female was decreased in females exposed to 42.5 ppm of arsenic in utero. In both 10-ppb and 42.5-ppm groups, arsenic-exposed females had significantly greater body weight gain, body fat content, and glucose intolerance. CONCLUSION: Our findings revealed unexpected effects of in utero exposure to arsenic: exposure to both a human-relevant low dose and a tumor-inducing level led to early onset of vaginal opening and to obesity in female CD-1 mice.


Asunto(s)
Arsenitos/toxicidad , Contaminantes Ambientales/toxicidad , Reproducción/efectos de los fármacos , Maduración Sexual/efectos de los fármacos , Compuestos de Sodio/toxicidad , Tejido Adiposo/efectos de los fármacos , Animales , Glucemia/metabolismo , Agua Potable/química , Ciclo Estral/efectos de los fármacos , Femenino , Fertilidad/efectos de los fármacos , Gonadotropinas/sangre , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Vagina/efectos de los fármacos , Vagina/fisiología
16.
Biol Reprod ; 90(3): 62, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24501173

RESUMEN

As the central component of canonical TGFbeta superfamily signaling, SMAD4 is a critical regulator of organ development, patterning, tumorigenesis, and many other biological processes. Because numerous TGFbeta superfamily ligands are expressed in developing testes, there may exist specific requirements for SMAD4 in individual testicular cell types. Previously, we reported that expansion of the fetal testis cords requires expression of SMAD4 by the Sertoli cell lineage. To further uncover the role of Smad4 in murine testes, we produced conditional knockout mice lacking Smad4 in either Leydig cells or in both Sertoli and Leydig cells simultaneously. Loss of Smad4 concomitantly in Sertoli and Leydig cells led to underdevelopment of the testis cords during fetal life and mild testicular dysgenesis in young adulthood (decreased testis size, partially dysgenic seminiferous tubules, and low sperm production). When the Sertoli/Leydig cell Smad4 conditional knockout mice aged (56- to 62-wk old), the testis phenotypes became exacerbated with the appearance of hemorrhagic tumors, Leydig cell adenomas, and a complete loss of spermatogenesis. In contrast, loss of Smad4 in Leydig cells alone did not appreciably alter fetal and adult testis development. Our findings support a cell type-specific requirement of Smad4 in testis development and suppression of testicular tumors.


Asunto(s)
Disgenesia Gonadal/genética , Disgenesia Gonadal/patología , Hemorragia/genética , Hemorragia/patología , Células Intersticiales del Testículo/fisiología , Células de Sertoli/fisiología , Proteína Smad4/genética , Proteína Smad4/fisiología , Neoplasias Testiculares/genética , Neoplasias Testiculares/patología , Adenoma/patología , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Hemorragia/etiología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , Túbulos Seminíferos/efectos de los fármacos , Túbulos Seminíferos/fisiología , Neoplasias Testiculares/complicaciones , Testículo/crecimiento & desarrollo , Testículo/fisiología
17.
FASEB J ; 27(7): 2657-66, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23568777

RESUMEN

Testes contain two distinct Leydig cell populations during development: fetal and adult Leydig cells (FLCs and ALCs, respectively). ALCs are not derived from FLCs, and it is unknown whether these two populations share common progenitors. We discovered that hedgehog (Hh) signaling is responsible for transforming steroidogenic factor 1-positive (SF1(+)) progenitors into FLCs. However, not all SF1(+) progenitors become FLCs, and some remain undifferentiated through fetal development. We therefore hypothesized that if FLCs and ALCs share SF1(+) progenitors, increased Hh pathway activation in SF1(+) progenitor cells could change the dynamics and distribution of SF1(+) progenitors, FLCs, and ALCs. Using a genetic model involving constitutive activation of Hh pathway in SF1(+) cells, we observed reduced numbers of SF1(+) progenitor cells and increased FLCs. Conversely, increased Hh activation led to decreased ALC populations prepubertally, while adult ALC numbers were comparable to control testes. Hence, reduction in SF1(+) progenitors temporarily affects ALC numbers, suggesting that SF1(+) progenitors in fetal testes are a potential source of both FLCs and ALCs. Besides transient ALC defects, adult animals with Hh activation in SF1(+) progenitors had reduced testicular weight, oligospermia, and decreased sperm mobility. These defects highlight the importance of properly regulated Hh signaling in Leydig cell development and testicular functions.


Asunto(s)
Proteínas Hedgehog/metabolismo , Células Intersticiales del Testículo/metabolismo , Células Madre/metabolismo , Factor Esteroidogénico 1/metabolismo , Factores de Edad , Animales , Recuento de Células , Diferenciación Celular , Proliferación Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Feto/citología , Feto/metabolismo , Proteínas Hedgehog/genética , Inmunohistoquímica , Células Intersticiales del Testículo/citología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Transducción de Señal , Motilidad Espermática , Células Madre/citología , Factor Esteroidogénico 1/genética , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Factores de Tiempo
18.
Mol Cell Endocrinol ; 361(1-2): 165-71, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22580128

RESUMEN

Functions of adrenal medulla, particularly synthesis of catecholamine, are under the control of glucocorticoids produced by the cortex. To further investigate whether development/differentiation of the adrenal medulla is associated with proper organization of the adrenal cortex, we examined development of the medulla in four different mouse models with various defects in the adrenal cortex. By using the Sf1/Cre mouse line that inactivates/activates genes in Steroidogenic factor 1 (SF1)-positive cells of the fetal adrenal cortex, we produced mice that exhibit either (1) cortex hypoplasia, (2) progressive degeneration of fetal adrenal cortex, (3) cortex dysgenesis, or (4) cortex-medulla disorganization. The formation of phenylethanolamine N-methyltransferase (PNMT)-positive medulla in all models indicates that differentiation of adrenal medulla is independent of the growth of adrenal cortex. However, the misplaced/dysgenic medulla in embryos where ß-catenin expression is altered, suggests that the ß-catenin pathway in the adrenal cortical cells plays an indirect role in controlling proper organization of the adrenal medulla.


Asunto(s)
Corteza Suprarrenal/patología , Médula Suprarrenal/patología , Diferenciación Celular , Corteza Suprarrenal/metabolismo , Médula Suprarrenal/metabolismo , Animales , ARN Helicasas DEAD-box/metabolismo , Proteínas Hedgehog/metabolismo , Integrasas/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Fenotipo , Ribonucleasa III/metabolismo , Factor Esteroidogénico 1/metabolismo , beta Catenina/deficiencia , beta Catenina/metabolismo
19.
Methods Mol Biol ; 825: 211-21, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22144247

RESUMEN

Genetic lineage tracing (or fate mapping) techniques are designed to permanently label progenitor cells of target tissues, thereby allowing delineation of the progenies of labeled cells during organogenesis. This technology has been widely used in the study of cell migration and lineage specification in various organs and organisms. Here, we describe how to apply the genetic lineage tracing model in combination with immunohistochemistry to identify the potential origins of somatic cell precursors in perinatal mouse ovaries.


Asunto(s)
Linaje de la Célula/genética , Inmunohistoquímica/métodos , Ovario/embriología , Animales , Cruzamiento/métodos , Evolución Clonal , Femenino , Técnicas de Genotipaje/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos/métodos , Organogénesis , Embarazo , Tamoxifeno/administración & dosificación
20.
Endocrinology ; 152(11): 4358-67, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21952240

RESUMEN

Proper development of the seminiferous tubules (or testis cords in embryos) is critical for male fertility. Sertoli cells, somatic components of the seminiferous tubules, serve as nurse cells to the male germline, and thus their numbers decide the quantity of sperm output in adulthood. We previously identified activin A, the protein product of the activin ßA (Inhba) gene, as a key regulator of murine Sertoli cell proliferation and testis cord expansion during embryogenesis. Although our genetic studies implicated fetal Leydig cells as the primary producers of testicular activin A, gonocytes are another potential source. To investigate the relative contribution of gonocyte-derived activin A to testis morphogenesis, we compared testis development in the Inhba global knockout mouse, which lacks activin A production in all cells (including the gonocytes), and a steroidogenic factor 1 (Sf1)-specific conditional knockout model in which activin A expression in testicular somatic cells is disrupted but gonocyte expression of activin A remains intact. Surprisingly, testis development was comparable in these two models of activin A insufficiency, with similar reductions in Sertoli cell proliferation and minor differences in testis histology. Thus, our findings suggest activin A from male gonocytes is insufficient to promote Sertoli cell proliferation and testis cord expansion in the absence of somatic cell-derived activin A. Evaluation of adult male mice with fetal disruption of activin A revealed reduced testis size, lowered sperm production, altered testicular histology, and elevated plasma FSH levels, defects reminiscent of human cases of androgen-sufficient idiopathic oligozoospermia.


Asunto(s)
Activinas/metabolismo , Células Intersticiales del Testículo/metabolismo , Morfogénesis/fisiología , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Testículo/embriología , Animales , Proliferación Celular , Hormona Folículo Estimulante/sangre , Masculino , Ratones , Ratones Transgénicos , Recuento de Espermatozoides , Testículo/crecimiento & desarrollo , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...