Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 150(24)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38078651

RESUMO

To investigate the role of the nuclear receptor NR5A1 in the testis after sex determination, we analyzed mice lacking NR5A1 in Sertoli cells (SCs) from embryonic day (E) 13.5 onwards. Ablation of Nr5a1 impaired the expression of genes characteristic of SC identity (e.g. Sox9 and Amh), caused SC death from E14.5 onwards through a Trp53-independent mechanism related to anoikis, and induced disorganization of the testis cords. Together, these effects caused germ cells to enter meiosis and die. Single-cell RNA-sequencing experiments revealed that NR5A1-deficient SCs changed their molecular identity: some acquired a 'pre-granulosa-like' cell identity, whereas other reverted to a 'supporting progenitor-like' cell identity, most of them being 'intersex' because they expressed both testicular and ovarian genes. Fetal Leydig cells (LCs) did not display significant changes, indicating that SCs are not required beyond E14.5 for their emergence or maintenance. In contrast, adult LCs were absent from postnatal testes. In addition, adult mutant males displayed persistence of Müllerian duct derivatives, decreased anogenital distance and reduced penis length, which could be explained by the loss of AMH and testosterone synthesis due to SC failure.


Assuntos
Anoikis , Células de Sertoli , Animais , Masculino , Camundongos , Anoikis/genética , Morte Celular/genética , Células de Sertoli/metabolismo , Testículo/metabolismo
2.
Development ; 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-38063846

RESUMO

To investigate the role of the nuclear receptor NR5A1 in testis after sex determination, we have analyzed mice lacking NR5A1 in Sertoli cells (SC) from embryonic day (E) 13.5 onwards. Ablation of Nr5a1 impairs the expression of genes characteristic of the SC identity (e.g., Sox9, Amh), causes SC death from E14.5 through a Trp53-independent mechanism related to anoikis, and induces disorganization of the testis cords. Together, these effects cause germ cells to enter meiosis and die. Single-cell RNA-sequencing experiments revealed that NR5A1-deficient SC change their molecular identity: some acquire a "pre-granulosa-like" identity, while other revert to a "supporting progenitor-like" cell identity, most of them being "intersex" because they express both testicular and ovarian genes. Fetal Leydig cells (LC) do not display significant changes, indicating that SC are not required beyond E14.5 for their emergence or maintenance. In contrast, adult LC were absent from the postnatal testes. In addition, adult mutant males display persistence of Müllerian duct derivatives, decreased anogenital distance and reduced penis length, which can be explained by the loss of AMH and testosterone synthesis due to SC failure.

3.
PLoS Biol ; 18(11): e3000902, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33201874

RESUMO

Coordinated development of muscles, tendons, and their attachment sites ensures emergence of functional musculoskeletal units that are adapted to diverse anatomical demands among different species. How these different tissues are patterned and functionally assembled during embryogenesis is poorly understood. Here, we investigated the morphogenesis of extraocular muscles (EOMs), an evolutionary conserved cranial muscle group that is crucial for the coordinated movement of the eyeballs and for visual acuity. By means of lineage analysis, we redefined the cellular origins of periocular connective tissues interacting with the EOMs, which do not arise exclusively from neural crest mesenchyme as previously thought. Using 3D imaging approaches, we established an integrative blueprint for the EOM functional unit. By doing so, we identified a developmental time window in which individual EOMs emerge from a unique muscle anlage and establish insertions in the sclera, which sets these muscles apart from classical muscle-to-bone type of insertions. Further, we demonstrate that the eyeballs are a source of diffusible all-trans retinoic acid (ATRA) that allow their targeting by the EOMs in a temporal and dose-dependent manner. Using genetically modified mice and inhibitor treatments, we find that endogenous local variations in the concentration of retinoids contribute to the establishment of tendon condensations and attachment sites that precede the initiation of muscle patterning. Collectively, our results highlight how global and site-specific programs are deployed for the assembly of muscle functional units with precise definition of muscle shapes and topographical wiring of their tendon attachments.


Assuntos
Músculos Oculomotores/embriologia , Músculos Oculomotores/crescimento & desenvolvimento , Tretinoína/metabolismo , Animais , Tecido Conjuntivo/fisiologia , Desenvolvimento Embrionário , Olho , Imageamento Tridimensional/métodos , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Morfogênese , Transdução de Sinais , Tendões/fisiologia , Tretinoína/fisiologia
4.
Development ; 146(13)2019 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-31273085

RESUMO

Retinoic acid (RA), a metabolite of retinol (vitamin A), functions as a ligand for nuclear RA receptors (RARs) that regulate development of chordate animals. RA-RARs can activate or repress transcription of key developmental genes. Genetic studies in mouse and zebrafish embryos that are deficient in RA-generating enzymes or RARs have been instrumental in identifying RA functions, revealing that RA signaling regulates development of many organs and tissues, including the body axis, spinal cord, forelimbs, heart, eye and reproductive tract. An understanding of the normal functions of RA signaling during development will guide efforts for use of RA as a therapeutic agent to improve human health. Here, we provide an overview of RA signaling and highlight its key functions during development.


Assuntos
Genes Controladores do Desenvolvimento , Receptores do Ácido Retinoico/fisiologia , Tretinoína/farmacologia , Tretinoína/fisiologia , Animais , Embrião de Mamíferos , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Controladores do Desenvolvimento/efeitos dos fármacos , Genes Controladores do Desenvolvimento/genética , Humanos , Camundongos , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Tretinoína/metabolismo , Peixe-Zebra
5.
Development ; 146(1)2019 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-30487180

RESUMO

In mammals, all-trans retinoic acid (ATRA) is instrumental to spermatogenesis. It is synthesized by two retinaldehyde dehydrogenases (RALDH) present in both Sertoli cells (SCs) and germ cells (GCs). In order to determine the relative contributions of each source of ATRA, we have generated mice lacking all RALDH activities in the seminiferous epithelium (SE). We show that both the SC- and GC-derived sources of ATRA cooperate to initiate and propagate spermatogenetic waves at puberty. In adults, they exert redundant functions and, against all expectations, the GC-derived source does not perform any specific roles despite contributing to two-thirds of the total amount of ATRA present in the testis. The production from SCs is sufficient to maintain the periodic expression of genes in SCs, as well and the cycle and wave of the SE, which account for the steady production of spermatozoa. The production from SCs is also specifically required for spermiation. Importantly, our study shows that spermatogonia differentiation depends upon the ATRA synthesized by RALDH inside the SE, whereas initiation of meiosis and expression of STRA8 by spermatocytes can occur without ATRA.


Assuntos
Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermatócitos/metabolismo , Espermatogênese/fisiologia , Espermatogônias/metabolismo , Tretinoína/metabolismo , Animais , Feminino , Masculino , Meiose/fisiologia , Camundongos , Camundongos Transgênicos , Epitélio Seminífero/citologia , Células de Sertoli/citologia , Espermatócitos/citologia , Espermatogônias/citologia
6.
Dev Biol ; 426(1): 17-27, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28456466

RESUMO

The differentiation of germ cells into oogonia or spermatogonia is the first step that eventually gives rise to fully mature gametes. In the female fetal gonad, the RSPO1/WNT/CTNNB1 signalling pathway is involved in primordial germ cell proliferation and differentiation into female germ cells, which are able to enter meiosis. In the postnatal testis, the WNT/CTNNB1 pathway also mediates proliferation of spermatogonial stem cells and progenitor cells. Here we show that forced activation of the WNT/CTNNB1 pathway in fetal gonocytes using transgenic mice leads to deregulated spermatogonial proliferation, and exhaustion of the spermatocytes by apoptosis, resulting in a hypoplastic testis. These findings demonstrate that a finely tuned timing in WNT/CTNNB1 signalling activity is required for spermatogenesis.


Assuntos
Células-Tronco Germinativas Adultas/citologia , Ativação Enzimática/fisiologia , Espermatogênese/fisiologia , Espermatogônias/citologia , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Espermatócitos/citologia , Testículo/patologia
7.
Dev Biol ; 424(2): 208-220, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28274610

RESUMO

Retinoic acid (RA) is a potent inducer of cell differentiation and plays an essential role in sex-specific germ cell development in the mammalian gonad. RA is essential for male gametogenesis and hence fertility. However, RA can also disrupt sexual cell fate in somatic cells of the testis, promoting transdifferentiation of male Sertoli cells to female granulosa-like cells when the male sexual regulator Dmrt1 is absent. The feminizing ability of RA in the Dmrt1 mutant somatic testis suggests that RA might normally play a role in somatic cell differentiation or cell fate maintenance in the ovary. To test for this possibility we disrupted RA signaling in somatic cells of the early fetal ovary using three genetic strategies and one pharmaceutical approach. We found that deleting all three RA receptors (RARs) in the XX somatic gonad at the time of sex determination did not significantly affect ovarian differentiation, follicle development, or female fertility. Transcriptome analysis of adult triple mutant ovaries revealed remarkably little effect on gene expression in the absence of somatic RAR function. Likewise, deletion of three RA synthesis enzymes (Aldh1a1-3) at the time of sex determination did not masculinize the ovary. A dominant-negative RAR transgene altered granulosa cell proliferation, likely due to interference with a non-RA signaling pathway, but did not prevent granulosa cell specification and oogenesis or abolish fertility. Finally, culture of fetal XX gonads with an RAR antagonist blocked germ cell meiotic initiation but did not disrupt sex-biased gene expression. We conclude that RA signaling, although crucial in the ovary for meiotic initiation, is not required for granulosa cell specification, differentiation, or reproductive function.


Assuntos
Ovário/embriologia , Ovário/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tretinoína/farmacologia , Família Aldeído Desidrogenase 1 , Animais , Linhagem da Célula/efeitos dos fármacos , Feminino , Feto/embriologia , Feto/metabolismo , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Inativação de Genes , Genes Dominantes , Isoenzimas/metabolismo , Masculino , Mamíferos , Meiose/efeitos dos fármacos , Mesonefro/efeitos dos fármacos , Mesonefro/embriologia , Mesonefro/metabolismo , Camundongos , Ovário/efeitos dos fármacos , Receptores do Ácido Retinoico/metabolismo , Retinal Desidrogenase/metabolismo , Retinoides/farmacologia , Processos de Determinação Sexual/efeitos dos fármacos , Técnicas de Cultura de Tecidos
8.
PLoS Genet ; 11(10): e1005501, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26427057

RESUMO

All-trans retinoic acid (ATRA) is instrumental to male germ cell differentiation, but its mechanism of action remains elusive. To address this question, we have analyzed the phenotypes of mice lacking, in spermatogonia, all rexinoid receptors (RXRA, RXRB and RXRG) or all ATRA receptors (RARA, RARB and RARG). We demonstrate that the combined ablation of RXRA and RXRB in spermatogonia recapitulates the set of defects observed both upon ablation of RAR in spermatogonia. We also show that ATRA activates RAR and RXR bound to a conserved regulatory region to increase expression of the SALL4A transcription factor in spermatogonia. Our results reveal that this major pluripotency gene is a target of ATRA signaling and that RAR/RXR heterodimers are the functional units driving its expression in spermatogonia. They add to the mechanisms through which ATRA promote expression of the KIT tyrosine kinase receptor to trigger a critical step in spermatogonia differentiation. Importantly, they indicate also that meiosis eventually occurs in the absence of a RAR/RXR pathway within germ cells and suggest that instructing this process is either ATRA-independent or requires an ATRA signal originating from Sertoli cells.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Proteínas Proto-Oncogênicas c-kit/genética , Espermatogônias/crescimento & desenvolvimento , Fatores de Transcrição/biossíntese , Tretinoína/metabolismo , Animais , Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Meiose/genética , Camundongos , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores X de Retinoides/genética , Células de Sertoli/metabolismo , Espermatogênese/genética , Espermatogônias/metabolismo , Fatores de Transcrição/genética
9.
Kidney Int ; 92(6): 1444-1457, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28756872

RESUMO

Proliferation of glomerular epithelial cells, including podocytes, is a key histologic feature of crescentic glomerulonephritis. We previously found that retinoic acid (RA) inhibits proliferation and induces differentiation of podocytes by activating RA receptor-α (RARα) in a murine model of HIV-associated nephropathy. Here, we examined whether RA would similarly protect podocytes against nephrotoxic serum-induced crescentic glomerulonephritis and whether this effect was mediated by podocyte RARα. RA treatment markedly improved renal function and reduced the number of crescentic lesions in nephritic wild-type mice, while this protection was largely lost in mice with podocyte-specific ablation of Rara (Pod-Rara knockout). At a cellular level, RA significantly restored the expression of podocyte differentiation markers in nephritic wild-type mice, but not in nephritic Pod-Rara knockout mice. Furthermore, RA suppressed the expression of cell injury, proliferation, and parietal epithelial cell markers in nephritic wild-type mice, all of which were significantly dampened in nephritic Pod-Rara knockout mice. Interestingly, RA treatment led to the coexpression of podocyte and parietal epithelial cell markers in a small subset of glomerular cells in nephritic mice, suggesting that RA may induce transdifferentiation of parietal epithelial cells toward a podocyte phenotype. In vitro, RA directly inhibited the proliferation of parietal epithelial cells and enhanced the expression of podocyte markers. In vivo lineage tracing of labeled parietal epithelial cells confirmed that RA increased the number of parietal epithelial cells expressing podocyte markers in nephritic glomeruli. Thus, RA attenuates crescentic glomerulonephritis primarily through RARα-mediated protection of podocytes and in part through the inhibition of parietal epithelial cell proliferation and induction of their transdifferentiation into podocytes.


Assuntos
Proliferação de Células/efeitos dos fármacos , Glomerulonefrite/tratamento farmacológico , Podócitos/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Receptor alfa de Ácido Retinoico/metabolismo , Tretinoína/farmacologia , Animais , Autoanticorpos/administração & dosagem , Autoanticorpos/imunologia , Biomarcadores/metabolismo , Biópsia , Cápsula Glomerular/citologia , Cápsula Glomerular/efeitos dos fármacos , Cápsula Glomerular/fisiologia , Transdiferenciação Celular/efeitos dos fármacos , Células Cultivadas , Glomerulonefrite/imunologia , Glomerulonefrite/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Podócitos/patologia , Podócitos/fisiologia , Substâncias Protetoras/uso terapêutico , Receptor alfa de Ácido Retinoico/genética , Tretinoína/uso terapêutico
10.
J Cell Sci ; 128(9): 1800-11, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25795304

RESUMO

The formation of programmed DNA double-strand breaks (DSBs) at the beginning of meiotic prophase marks the initiation of meiotic recombination. Meiotic DSB formation is catalyzed by SPO11 and their repair takes place on meiotic chromosome axes. The evolutionarily conserved MEI4 protein is required for meiotic DSB formation and is localized on chromosome axes. Here, we show that HORMAD1, one of the meiotic chromosome axis components, is required for MEI4 localization. Importantly, the quantitative correlation between the level of axis-associated MEI4 and DSB formation suggests that axis-associated MEI4 could be a limiting factor for DSB formation. We also show that MEI1, REC8 and RAD21L are important for proper MEI4 localization. These findings on MEI4 dynamics during meiotic prophase suggest that the association of MEI4 to chromosome axes is required for DSB formation, and that the loss of this association upon DSB repair could contribute to turning off meiotic DSB formation.


Assuntos
Quebras de DNA de Cadeia Dupla , Meiose , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Pareamento Cromossômico , Cromossomos de Mamíferos/metabolismo , Prófase Meiótica I , Camundongos , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Subunidades Proteicas/metabolismo , Transporte Proteico , Fatores de Tempo , Coesinas
11.
Exp Eye Res ; 154: 190-195, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27840061

RESUMO

Retinoic acid (RA) is a biologically active metabolite of vitamin A (retinol) that serves as an important signaling molecule in orchestrating diverse developmental processes including multiple roles during ocular development. Loss-of-function studies using gene knockouts of RA-synthesizing enzymes encoded by Aldh1a1, Aldh1a2, and Aldh1a3 (also known as Raldh1, Raldh2, and Raldh3) have provided valuable insight into how RA controls eye morphogenesis including corneal development. However, it is unclear whether endogenous RA is required for maintenance and regeneration of adult cornea. Here, we investigated the role of Aldh1a genes in the adult cornea using a novel conditional Aldh1a1,2,3-flox/flox;Rosa26-CreERT2 loss-of-function mouse model to determine the biological function of RA. Our findings indicate that loss of RA synthesis results in corneal thinning characterized by reduced thickness of the stromal layer, impaired corneal epithelial cell proliferation, and increased apoptosis. Corneal thinning in Aldh1a-deficient mice was significantly rescued by RA administration, indicating an important role of endogenous RA signaling in adult corneal homeostasis and regeneration. Thus, Aldh1a1,2,3-flox/flox;Rosa26-CreERT2 mice provide a useful model for investigating the mechanistic role of RA signaling in adult corneal maintenance and could provide new insights into therapeutic approaches for controlling corneal repair to prevent vision loss.


Assuntos
Apoptose , Epitélio Corneano/metabolismo , Regeneração/fisiologia , Tretinoína/metabolismo , Animais , Proliferação de Células , Epitélio Corneano/patologia , Camundongos , Transdução de Sinais
12.
Immunity ; 29(5): 758-70, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-19006694

RESUMO

CD4(+)Foxp3(+) regulatory T (Treg) cells originate primarily from thymic differentiation, but conversion of mature T lymphocytes to Foxp3 positivity can be elicited by several means, including in vitro activation in the presence of TGF-beta. Retinoic acid (RA) increases TGF-beta-induced expression of Foxp3, through unknown molecular mechanisms. We showed here that, rather than enhancing TGF-beta signaling directly in naive CD4(+) T cells, RA negatively regulated an accompanying population of CD4(+) T cells with a CD44(hi) memory and effector phenotype. These memory cells actively inhibited the TGF-beta-induced conversion of naive CD4(+) T cells through the synthesis of a set of cytokines (IL-4, IL-21, IFN-gamma) whose expression was coordinately curtailed by RA. This indirect effect was evident in vivo and required the expression of the RA receptor alpha. Thus, cytokine-producing CD44(hi) cells actively restrain TGF-beta-mediated Foxp3 expression in naive T cells, and this balance can be shifted or fine-tuned by RA.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fatores de Transcrição Forkhead/metabolismo , Receptores do Ácido Retinoico/imunologia , Linfócitos T Reguladores/imunologia , Tretinoína/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Homeostase , Receptores de Hialuronatos/análise , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-4/imunologia , Interleucina-4/metabolismo , Interleucinas/imunologia , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores do Ácido Retinoico/deficiência , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
13.
Biochim Biophys Acta ; 1849(2): 84-93, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24875094

RESUMO

All-trans retinoic acid (atRA), the active metabolite of vitamin A, plays critical functions in spermatogenesis, a complex, highly organized and regulated process comprising three phases. During the proliferative phase, undifferentiated spermatogonia divide to maintain a stem cell population and expand a progenitor cell population, of which a fraction enters the differentiation pathway yielding primary spermatocytes. During the meiotic phase, primary spermatocytes undergo recombination, segregation and reduction by half of chromosomes to produce haploid round spermatids. During the morphogenetic, post-meiotic phase, spermatids differentiate and elongate to ultimately form spermatozoa. Studies performed during the past 20 years have significantly improved our knowledge on the location of the proteins transducing the atRA signal, on the target genes of atRA and on its mechanism of action. This article is part of a Special Issue entitled: Nuclear receptors in animal development.


Assuntos
Receptores do Ácido Retinoico/fisiologia , Espermatogênese , Espermatogônias/citologia , Animais , Masculino , Células de Sertoli/citologia , Células de Sertoli/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Tretinoína/farmacologia
14.
PLoS Genet ; 8(12): e1003170, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300469

RESUMO

In mammals, male sex determination is governed by SRY-dependent activation of Sox9, whereas female development involves R-spondin1 (RSPO1), an activator of the WNT/beta-catenin signaling pathway. Genetic analyses in mice have demonstrated Sry and Sox9 to be both required and sufficient to induce testicular development. These genes are therefore considered as master regulators of the male pathway. Indeed, female-to-male sex reversal in XX Rspo1 mutant mice correlates with Sox9 expression, suggesting that this transcription factor induces testicular differentiation in pathological conditions. Unexpectedly, here we show that testicular differentiation can occur in XX mutants lacking both Rspo1 and Sox9 (referred to as XX Rspo1(KO)Sox9(cKO) ()), indicating that Sry and Sox9 are dispensable to induce female-to-male sex reversal. Molecular analyses show expression of both Sox8 and Sox10, suggesting that activation of Sox genes other than Sox9 can induce male differentiation in Rspo1(KO)Sox9(cKO) mice. Moreover, since testis development occurs in XY Rspo1(KO)Sox9(cKO) mice, our data show that Rspo1 is the main effector for male-to-female sex reversal in XY Sox9(cKO) mice. Thus, Rspo1 is an essential activator of ovarian development not only in normal situations, but also in sex reversal situations. Taken together these data demonstrate that both male and female sex differentiation is induced by distinct, active, genetic pathways. The dogma that considers female differentiation as a default pathway therefore needs to be definitively revised.


Assuntos
Fatores de Transcrição SOX9/genética , Processos de Determinação Sexual/genética , Diferenciação Sexual/genética , Testículo , Trombospondinas/genética , Animais , Diferenciação Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes sry , Humanos , Masculino , Camundongos , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais , Testículo/citologia , Testículo/crescimento & desenvolvimento , Trombospondinas/metabolismo , Via de Sinalização Wnt
15.
Proc Natl Acad Sci U S A ; 109(41): 16582-7, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23012458

RESUMO

Direct evidence for a role of endogenous retinoic acid (RA), the active metabolite of vitamin A in the initial differentiation and meiotic entry of spermatogonia, and thus in the initiation of spermatogenesis is still lacking. RA is synthesized by dedicated enzymes, the retinaldehyde dehydrogenases (RALDH), and binds to and activates nuclear RA receptors (RARA, RARB, and RARG) either within the RA-synthesizing cells or in the neighboring cells. In the present study, we have used a combination of somatic genetic ablations and pharmacological approaches in vivo to show that during the first, prepubertal, spermatogenic cycle (i) RALDH-dependent synthesis of RA by Sertoli cells (SC), the supporting cells of the germ cell (GC) lineage, is indispensable to initiate differentiation of A aligned into A1 spermatogonia; (ii) RARA in SC mediates the effects of RA, possibly through activating Mafb expression, a gene whose Drosophila homolog is mandatory to GC differentiation; (iii) RA synthesized by premeiotic spermatocytes cell autonomously induces meiotic initiation through controlling the RAR-dependent expression of Stra8. Furthermore, we show that RA of SC origin is no longer necessary for the subsequent spermatogenic cycles but essential to spermiation. Altogether, our data establish that the effects of RA in vivo on spermatogonia differentiation are indirect, via SC, but direct on meiotic initiation in spermatocytes, supporting thereby the notion that, contrary to the situation in the female, RA is necessary to induce meiosis in the male.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Meiose/efeitos dos fármacos , Células de Sertoli/efeitos dos fármacos , Espermatócitos/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Tretinoína/farmacologia , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Diferenciação Celular/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Meiose/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/genética , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Retinal Desidrogenase/genética , Retinal Desidrogenase/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Sertoli/metabolismo , Espermatócitos/metabolismo , Espermatogônias/citologia , Espermatogônias/metabolismo , Tretinoína/metabolismo
16.
J Biol Chem ; 288(34): 24528-39, 2013 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-23839944

RESUMO

The plasma membrane protein STRA6 is thought to mediate uptake of retinol from its blood carrier retinol-binding protein (RBP) into cells and to function as a surface receptor that, upon binding of holo-RBP, activates a JAK/STAT cascade. It was suggested that STRA6 signaling underlies insulin resistance induced by elevated serum levels of RBP in obese animals. To investigate these activities in vivo, we generated and analyzed Stra6-null mice. We show that the contribution of STRA6 to retinol uptake by tissues in vivo is small and that, with the exception of the eye, ablation of Stra6 has only a modest effect on retinoid homeostasis and does not impair physiological functions that critically depend on retinoic acid in the embryo or in the adult. However, ablation of Stra6 effectively protects mice from RBP-induced suppression of insulin signaling. Thus one biological function of STRA6 in tissues other than the eye appears to be the coupling of circulating holo-RBP levels to cell signaling, in turn regulating key processes such as insulin response.


Assuntos
Resistência à Insulina , Insulina/metabolismo , Proteínas de Membrana/metabolismo , Obesidade/metabolismo , Proteínas Plasmáticas de Ligação ao Retinol/metabolismo , Vitamina A/metabolismo , Células 3T3-L1 , Animais , Olho , Insulina/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Obesidade/genética , Obesidade/patologia , Proteínas Plasmáticas de Ligação ao Retinol/genética , Transdução de Sinais/genética
17.
Development ; 138(13): 2661-72, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21613325

RESUMO

Mitosis is controlled by multiple kinases that drive cell cycle progression and prevent chromosome mis-segregation. Aurora kinase B interacts with survivin, borealin and incenp to form the chromosomal passenger complex (CPC), which is involved in the regulation of microtubule-kinetochore attachments and cytokinesis. Whereas genetic ablation of survivin, borealin or incenp results in early lethality at the morula stage, we show here that aurora B is dispensable for CPC function during early cell divisions and aurora B-null embryos are normally implanted. This is due to a crucial function of aurora C during these early embryonic cycles. Expression of aurora C decreases during late blastocyst stages resulting in post-implantation defects in aurora B-null embryos. These defects correlate with abundant prometaphase figures and apoptotic cell death of the aurora B-deficient inner cell mass. Conditional deletion of aurora B in somatic cells that do not express aurora C results in chromosomal misalignment and lack of chromosome segregation. Re-expression of wild-type, but not kinase-dead, aurora C rescues this defect, suggesting functional overlap between these two kinases. Finally, aurora B-null cells partially arrest in the presence of nocodazole, suggesting that this kinase is not essential for the spindle assembly checkpoint.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Animais , Aurora Quinase B , Aurora Quinase C , Aurora Quinases , Blastocisto/metabolismo , Divisão Celular/genética , Divisão Celular/fisiologia , Células Cultivadas , Segregação de Cromossomos/genética , Segregação de Cromossomos/fisiologia , Feminino , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos , Camundongos Transgênicos , Mitose/genética , Mitose/fisiologia , Gravidez , Proteínas Serina-Treonina Quinases/genética , Fuso Acromático/genética , Fuso Acromático/metabolismo , Zigoto/metabolismo
18.
Biomedicines ; 11(1)2023 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-36672706

RESUMO

It has been established for almost 30 years that the retinoic acid receptor (RAR) signalling pathway plays essential roles in the morphogenesis of a large variety of organs and systems. Here, we used a temporally controlled genetic ablation procedure to precisely determine the time windows requiring RAR functions. Our results indicate that from E8.5 to E9.5, RAR functions are critical for the axial rotation of the embryo, the appearance of the sinus venosus, the modelling of blood vessels, and the formation of forelimb buds, lung buds, dorsal pancreatic bud, lens, and otocyst. They also reveal that E9.5 to E10.5 spans a critical developmental period during which the RARs are required for trachea formation, lung branching morphogenesis, patterning of great arteries derived from aortic arches, closure of the optic fissure, and growth of inner ear structures and of facial processes. Comparing the phenotypes of mutants lacking the 3 RARs with that of mutants deprived of all-trans retinoic acid (ATRA) synthesising enzymes establishes that cardiac looping is the earliest known morphogenetic event requiring a functional ATRA-activated RAR signalling pathway.

19.
Dev Cell ; 13(2): 242-53, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17681135

RESUMO

Integrity and preservation of a transparent cornea are essential for good vision. The corneal epithelium is stratified and nonkeratinized and is maintained and repaired by corneal stem cells. Here we demonstrate that Notch1 signaling is essential for cell fate maintenance of corneal epithelium during repair. Inducible ablation of Notch1 in the cornea combined with mechanical wounding show that Notch1-deficient corneal progenitor cells differentiate into a hyperplastic, keratinized, skin-like epithelium. This cell fate switch leads to corneal blindness and involves cell nonautonomous processes, characterized by secretion of fibroblast growth factor-2 (FGF-2) through Notch1(-/-) epithelium followed by vascularization and remodeling of the underlying stroma. Vitamin A deficiency is known to induce a similar corneal defect in humans (severe xerophthalmia). Accordingly, we found that Notch1 signaling is linked to vitamin A metabolism by regulating the expression of cellular retinol binding protein 1 (CRBP1), required to generate a pool of intracellular retinol.


Assuntos
Linhagem da Célula , Epitélio Corneano/citologia , Receptor Notch1/metabolismo , Transdução de Sinais , Vitamina A/metabolismo , Cicatrização , Animais , Diferenciação Celular , Movimento Celular , Substância Própria/patologia , Células Epidérmicas , Epitélio Corneano/patologia , Queratinas/metabolismo , Glândulas Tarsais/anormalidades , Camundongos , Modelos Biológicos , Receptor Notch1/deficiência , Proteínas de Ligação ao Retinol/metabolismo , Proteínas Celulares de Ligação ao Retinol , Células-Tronco/citologia
20.
Proc Natl Acad Sci U S A ; 106(11): 4272-7, 2009 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19255444

RESUMO

Retinoic acid (RA) receptors (RARs) alpha, beta, and gamma heterodimerized with rexinoid receptors (RXRs) alpha, beta, and gamma mediate the RA signal. To analyze the contribution of the transcriptional activity of RXRalpha, the main RXR during embryogenesis, we have engineered a mouse line harboring a transcriptionally silent RXRalpha mutant that lacks the activation functions AF1 and AF2. All homozygous mutants (Rxra(afo)) display the ocular defects previously observed in compound Rar-null and Rxra/Rar-null mutants, thus demonstrating that a transcriptionally active RXRalpha is required during eye development. In contrast, the vast majority of Rxra(afo) fetuses do not display the Rxra-null mutant hypoplasia of the myocardium, thus demonstrating that RXRalpha can act as a transcriptionally silent heterodimerization partner. Similarly, a transcriptionally silent RXRalpha mutant can support early embryogenesis, as Rxra(afo)/Rxrb-null embryos display a normal morphology, contrasting with the severe malformations exhibited by compound Rxra/Rxrb-null embryos. Along the same line, we show that a silent RXRalpha mutant is sufficient to allow the initial formation of the placental labyrinth, whereas later steps of trophoblast cell differentiation critically requires the AF2, but not the AF1, function of RXRalpha.


Assuntos
Morfogênese/genética , Receptores do Ácido Retinoico/fisiologia , Transcrição Gênica , Animais , Embrião de Mamíferos , Olho/embriologia , Olho/crescimento & desenvolvimento , Feminino , Genótipo , Coração/embriologia , Coração/crescimento & desenvolvimento , Camundongos , Camundongos Mutantes , Placenta/citologia , Gravidez , Receptores do Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Trofoblastos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA