Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Biol Reprod ; 110(6): 1055-1064, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38315794

RESUMO

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.


Assuntos
Técnicas de Cultura de Órgãos , Animais , Técnicas de Cultura de Órgãos/métodos , Camundongos , Masculino , Reprodução/fisiologia , Uretra , Pênis , Feminino
2.
Reproduction ; 168(5)2024 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-39190000

RESUMO

In brief: Female hypospadias is a little-known and poorly studied birth defect. This research establishes an anatomical and molecular foundation for future research to investigate the origins of this defect. Abstract: Hypospadias is a congenital anomaly of the external genitalia where the urethra does not properly close. In humans, hypospadias is mostly reported in male newborns, whereas in females hypospadias is rare, although it is generally considered to be under-reported. Improper urethra closure in the female genitalia can cause recurrent genitourinary tract infections and infertility. In mice, female hypospadias was induced by exposure to exogenous estrogenic compounds. Aside from the link between estrogen exposure and female hypospadias, the process of female urethra closure is largely unstudied, with the precise timing of urethra closure and associated molecular mechanisms remaining poorly understood. To address this gap, we determined when urethra closure occurs and identified gene expression patterns during the process of urethra closure in female neonatal mice from postnatal day (PND) 5 to 10. Using whole mount imaging and histology, we discovered that the initiation of urethra closure begins at PND7, and urethra closure is fully completed by PND10. To identify the genes associated with urethra closure, we conducted bulk RNA sequencing on female external genitalia prior to and after urethra closure. Gene ontology analyses revealed an increase in steroidogenic gene expression (Star, Hsd3b6, and Cyp17a1) during urethra closure, suggesting that the female genitalia locally produce steroids which could facilitate steroid signaling within the genitalia. With this study, we establish an anatomical timeline of female urethra closure and hypothesize a paracrine steroid signaling mechanism of urethra closure. These observations provide entry points to aid in further understanding external genital abnormalities, like hypospadias, in females.


Assuntos
Animais Recém-Nascidos , Hipospadia , Uretra , Animais , Feminino , Uretra/patologia , Camundongos , Hipospadia/patologia , Hipospadia/genética , Masculino , Camundongos Endogâmicos C57BL , Regulação da Expressão Gênica no Desenvolvimento
3.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34155146

RESUMO

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.


Assuntos
Genitália/citologia , Genitália/embriologia , Caracteres Sexuais , Análise de Célula Única , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hormônios/metabolismo , Masculino , Mesoderma/citologia , Mesoderma/embriologia , Camundongos Endogâmicos C57BL , Modelos Biológicos
4.
Biol Reprod ; 108(6): 866-870, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-36951956

RESUMO

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.


Assuntos
Freemartinismo , Testículo , Bovinos , Masculino , Feminino , Animais , Camundongos , Testículo/metabolismo , Freemartinismo/metabolismo , Sêmen , Células de Sertoli/metabolismo , Células Intersticiais do Testículo/metabolismo
5.
FASEB J ; 35(8): e21770, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34288113

RESUMO

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.


Assuntos
Fertilidade , Infertilidade/fisiopatologia , Obesidade/fisiopatologia , Ovário/fisiopatologia , Fator Esteroidogênico 1/fisiologia , Animais , Feminino , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
6.
FASEB J ; 35(9): e21876, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34449112

RESUMO

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.


Assuntos
Galinhas/genética , Feminização/genética , Gônadas/fisiologia , Fatores de Transcrição/genética , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Redes Reguladoras de Genes/genética , Masculino , Ovário/fisiologia , Cromossomos Sexuais , Testículo/fisiologia
7.
Biol Reprod ; 101(3): 602-616, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31058957

RESUMO

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.


Assuntos
Pesquisa Biomédica , Genitália/embriologia , Morfogênese/fisiologia , Diferenciação Sexual/fisiologia , Adulto , Animais , Pesquisa Biomédica/história , Pesquisa Biomédica/métodos , Pesquisa Biomédica/tendências , Feminino , Regulação da Expressão Gênica no Desenvolvimento , História do Século XX , História do Século XXI , Humanos , Invenções , Masculino , Reprodução/genética , Sistema Urogenital/embriologia
8.
Biol Reprod ; 97(3): 365-377, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-29088396

RESUMO

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.


Assuntos
Biologia Computacional , Simulação por Computador , Ovário/crescimento & desenvolvimento , Animais , Adesão Celular , Movimento Celular , Desenvolvimento Embrionário/fisiologia , Feminino , Células Germinativas , Células da Granulosa/fisiologia , Camundongos , Mitose , Método de Monte Carlo , Ovário/embriologia , Gravidez , Diferenciação Sexual , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Software , Fator de Células-Tronco
9.
Biol Reprod ; 90(3): 62, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24501173

RESUMO

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.


Assuntos
Disgenesia Gonadal/genética , Disgenesia Gonadal/patologia , Hemorragia/genética , Hemorragia/patologia , Células Intersticiais do Testículo/fisiologia , Células de Sertoli/fisiologia , Proteína Smad4/genética , Proteína Smad4/fisiologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia , Adenoma/patologia , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Hemorragia/etiologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Túbulos Seminíferos/efeitos dos fármacos , Túbulos Seminíferos/fisiologia , Neoplasias Testiculares/complicações , Testículo/crescimento & desenvolvimento , Testículo/fisiologia
10.
FASEB J ; 27(7): 2657-66, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23568777

RESUMO

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.


Assuntos
Proteínas Hedgehog/metabolismo , Células Intersticiais do Testículo/metabolismo , Células-Tronco/metabolismo , Fator Esteroidogênico 1/metabolismo , Fatores Etários , Animais , Contagem de Células , Diferenciação Celular , Proliferação de Células , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Feto/citologia , Feto/metabolismo , Proteínas Hedgehog/genética , Imuno-Histoquímica , Células Intersticiais do Testículo/citologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Transdução de Sinais , Motilidade dos Espermatozoides , Células-Tronco/citologia , Fator Esteroidogênico 1/genética , Testículo/citologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Fatores de Tempo
11.
bioRxiv ; 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39345510

RESUMO

Testicular fetal Leydig cells are a specialized cell type responsible for embryo masculinization. Fetal Leydig cells produce androgens, that induce the differentiation of male reproductive system and sexual characteristics. Deficiencies in Leydig cell differentiation leads to various disorders of sex development and male reproductive defects such as ambiguous genitalia, hypospadias, cryptorchidism, and infertility. Fetal Leydig cells are thought to originate from proliferating progenitor cells in the testis interstitium, marked by genes like Arx , Pdgfra , Tcf21 and Wnt5a . However, the precise mechanisms governing the transition from interstitial cells to fetal Leydig cells remain elusive. Through integrated approaches involving mouse models and single-nucleus multiomic analyses, we discovered that fetal Leydig cells originate from a Nr2f2 -positive non-steroidogenic interstitial cell population. Embryonic deletion of Nr2f2 in mouse testes resulted in disorders of sex development, including dysgenic testes, Leydig cell hypoplasia, cryptorchidism, and hypospadias. We found that NR2F2 promotes the progenitor cell fate while suppresses Leydig cell differentiation by directly and indirectly controlling a cohort of transcription factors and downstream genes. Bioinformatic analyses of single-nucleus ATAC-seq and NR2F2 ChIP-seq data revealed putative transcription factors co-regulating the process of interstitial to Leydig cell differentiation. Collectively, our findings not only highlight the critical role of Nr2f2 in orchestrating the transition from interstitial cells to fetal Leydig cells, but also provide molecular insight into the disorders of sex development as a result of Nr2f2 mutations.

12.
bioRxiv ; 2024 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-39386556

RESUMO

Accurate specification of female and male germ cells during embryonic development is critical for sexual reproduction. Primordial germ cells (PGCs) are the bipotential precursors of mature gametes that commit to an oogenic or spermatogenic fate in response to sex-determining cues from the fetal gonad. The critical processes required for PGCs to integrate and respond to signals from the somatic environment in gonads are not understood. In this study, we developed the first single-nucleus multiomics map of chromatin accessibility and gene expression during murine PGC development in both XX and XY embryos. Profiling of cell-type specific transcriptomes and regions of open chromatin from the same cell captured the molecular signatures and gene networks underlying PGC sex determination. Joint RNA and ATAC data for single PGCs resolved previously unreported PGC subpopulations and cataloged a multimodal reference atlas of differentiating PGC clusters. We discovered that regulatory element accessibility precedes gene expression during PGC development, suggesting that changes in chromatin accessibility may prime PGC lineage commitment prior to differentiation. Similarly, we found that sexual dimorphism in chromatin accessibility and gene expression increased temporally in PGCs. Combining single-nucleus sequencing data, we computationally mapped the cohort of transcription factors that regulate the expression of sexually dimorphic genes in PGCs. For example, the gene regulatory networks of XX PGCs are enriched for the transcription factors, TFAP2c, TCFL5, GATA2, MGA, NR6A1, TBX4, and ZFX. Sex-specific enrichment of the forkhead-box and POU6 families of transcription factors was also observed in XY PGCs. Finally, we determined the temporal expression patterns of WNT, BMP, and RA signaling during PGC sex determination, and our discovery analyses identified potentially new cell communication pathways between supporting cells and PGCs. Our results illustrate the diversity of factors involved in programming PGCs towards a sex-specific fate.

13.
Proc Natl Acad Sci U S A ; 107(23): 10526-31, 2010 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-20498064

RESUMO

Formation of tubular structures relies upon complex interactions between adjacent epithelium and mesenchyme. In the embryonic testes, dramatic compartmentalization leads to the formation of testis cords (epithelium) and the surrounding interstitium (mesenchyme). Sertoli cells, the epithelial cell type within testis cords, produce signaling molecules to orchestrate testis cord formation. The interstitial fetal Leydig cells, however, are thought only to masculinize the embryo and are not known to be involved in testis cord morphogenesis. Contrary to this notion, we have identified activin A, a member of the TGF-beta protein superfamily, as a product of the murine fetal Leydig cells that acts directly upon Sertoli cells to promote their proliferation during late embryogenesis. Genetic disruption of activin betaA, the gene encoding activin A, specifically in fetal Leydig cells resulted in a failure of fetal testis cord elongation and expansion due to decreased Sertoli cell proliferation. Conditional inactivation of Smad4, the central component of TGF-beta signaling, in Sertoli cells led to testis cord dysgenesis and proliferative defects similar to those of Leydig cell-specific activin betaA knockout testes. These results indicate that activin A is the major TGF-beta protein that acts directly on Sertoli cells. Testicular dysgenesis in activin betaA and Smad4 conditional knockout embryos persists into adulthood, leading to low sperm production and abnormal testicular histology. Our findings challenge the paradigm that fetal testis development is solely under the control of Sertoli cells, by uncovering an active and essential role of fetal Leydig cells during testis cord morphogenesis.


Assuntos
Subunidades beta de Inibinas/metabolismo , Células Intersticiais do Testículo/metabolismo , Comunicação Parácrina , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Testículo/embriologia , Testículo/metabolismo , Animais , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Subunidades beta de Inibinas/genética , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Transdução de Sinais , Proteína Smad4/metabolismo
14.
bioRxiv ; 2023 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-37986842

RESUMO

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.

15.
PNAS Nexus ; 1(4): pgac182, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36204418

RESUMO

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.

16.
Nat Commun ; 13(1): 4130, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35840551

RESUMO

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.


Assuntos
Ativinas , Hormônio Antimülleriano , Diferenciação Celular , Testículo , Ativinas/metabolismo , Ativinas/farmacologia , Animais , Hormônio Antimülleriano/metabolismo , Hormônio Antimülleriano/farmacologia , Comunicação Autócrina/efeitos dos fármacos , Comunicação Autócrina/fisiologia , Diferenciação Celular/fisiologia , Feminino , Masculino , Mamíferos , Camundongos , Comunicação Parácrina/fisiologia , Sêmen , Células de Sertoli , Testículo/metabolismo
17.
Mol Reprod Dev ; 77(6): 489-96, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20422709

RESUMO

Adrenal, testis, and ovary are steroidogenic organs derived from a common primordium that consists of steroidogenic factor 1 (SF1)-positive precursor cells. SF1 not only defines the steroidogenic lineages in these organs but also controls their differentiation. Recent evidence implicates the Hedgehog (Hh) signaling pathway as a downstream regulator of SF1 in the appearance of steroidogenic cells in these organs. The Hh signaling pathway serves as a common crosstalk component, yet has evolved diverse functions in the expansion and differentiation of the steroidogenic cells in a tissue-specific manner. The purpose of this review is to compare and contrast the different roles of Hh signaling in these three organs during development.


Assuntos
Glândulas Suprarrenais , Proteínas Hedgehog/metabolismo , Ovário , Transdução de Sinais/fisiologia , Testículo , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/embriologia , Glândulas Suprarrenais/metabolismo , Animais , Linhagem da Célula , Feminino , Proteínas Hedgehog/genética , Masculino , Ovário/citologia , Ovário/embriologia , Ovário/metabolismo , Testículo/citologia , Testículo/embriologia , Testículo/metabolismo
18.
Reprod Toxicol ; 95: 59-65, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32416200

RESUMO

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.


Assuntos
Retardadores de Chama/toxicidade , Bifenil Polibromatos/toxicidade , Animais , Feminino , Masculino , Troca Materno-Fetal , Gravidez , Próstata/efeitos dos fármacos , Ratos Wistar , Saúde Reprodutiva , Contagem de Espermatozoides , Espermatozoides/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/metabolismo
19.
Genesis ; 47(1): 40-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18979542

RESUMO

Interactions between adjacent epithelial and mesenchymal tissues represent a highly conserved mechanism in embryonic organogenesis. In particular, the ability of the mesenchyme to instruct cellular differentiation of the epithelium is a fundamental requirement for the morphogenesis of tubular structures such as those found in the kidneys, lungs, and the developing male reproductive system. Once the tubular structure has formed, it receives signals from the mesenchyme, which can control proliferation, patterning, and differentiation of the epithelium inside the tube. However, the epithelium is not a "silent partner" in this process, and epithelium-derived factors are often required for proper maintenance of the mesenchymal compartment. Although much emphasis has been placed on the characterization of mesenchymally-derived signals required for epithelial differentiation, it is important to note that epithelial-mesenchymal interactions are a two-way street wherein each compartment requires the presence of the other for proper tubule morphogenesis and function. In this review, we discuss epithelial-mesenchymal interactions in the processes of Wolffian duct and fetal testis cord development using the mouse as a model organism and propose inhibin beta A as a conserved mesenchyme-derived regulator in these two male-specific tubular structures.


Assuntos
Comunicação Celular , Epitélio/embriologia , Epitélio/metabolismo , Mesoderma/metabolismo , Testículo/embriologia , Testículo/metabolismo , Ductos Mesonéfricos/metabolismo , Animais , Diferenciação Celular , Masculino , Mesoderma/citologia , Mesoderma/embriologia , Testículo/citologia , Ductos Mesonéfricos/citologia , Ductos Mesonéfricos/embriologia
20.
Endocr Rev ; 39(5): 739-759, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29771299

RESUMO

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.


Assuntos
Linhagem da Célula/fisiologia , Células da Granulosa/citologia , Ovário/citologia , Células de Sertoli/citologia , Testículo/citologia , Animais , Diferenciação Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Feminino , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA