Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
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
2.
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
3.
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
4.
Proc Natl Acad Sci U S A ; 105(7): 2598-603, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18287084

RESUMO

Calcification of arteries is a major risk factor for cardiovascular mortality in humans. Using genetic approaches, we demonstrate here that the transcriptional intermediary factor 1alpha (TIF1alpha), recently shown to function as a tumor suppressor in murine hepatocytes, also participates in a molecular cascade that prevents calcifications in arterioles and medium-sized arteries. We further provide genetic evidence that this function of TIF1alpha is not exerted in hepatocytes. The sites of ectopic calcifications in mutant mice lacking TIF1alpha resemble those seen in mice carrying an activating mutation of the calcium sensor receptor (Casr) gene and, in TIF1alpha-deficient kidneys, Casr expression is increased together with that of many other vitamin D receptor (VDR) direct target genes, namely Car2, Cyp24a1, Trpv5, Trpv6, Calb1, S100g, Pthlh, and Spp1. Thus, our data indicate that TIF1alpha represses the VDR pathway in kidney and suggest that an up-regulation of Casr expression in this organ could account for ectopic calcifications generated upon TIF1alpha deficiency. Interestingly, the calcifying arteriopathy of TIF1alpha-null mutant mice shares features with the human age-related Mönckeberg's disease and, overall, the TIF1alpha-null mutant pathological phenotype supports the hypothesis that aging is promoted by increased activity of the vitamin D signaling pathway.


Assuntos
Artérias/metabolismo , Calcinose/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/metabolismo , Receptores de Calcitriol/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/metabolismo , Envelhecimento/fisiologia , Animais , Calcinose/genética , Cálcio/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Homeostase , Rim/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Vibrissas/metabolismo
5.
Reproduction ; 136(5): 619-26, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18713813

RESUMO

Somatic, targeted inactivation of the retinoid X receptor beta gene (Rxrb) in Sertoli cells (SC; yielding Rxrb(Ser-/-) mutants) leads to failure of spermatid release, accumulation of cholesterol esters and, subsequently, testis degeneration. These abnormalities are identical, in their nature and kinetics, to those observed upon inactivating Rxrb in the whole organism, thereby demonstrating that all reproductive functions of RXRB are carried out in SC. The Rxrb(Ser-/-) testis degeneration is a consequence of a cholesterol ester cell overload occurring in SC in response to reduced ABCA1- and SCARB1-mediated cholesterol efflux. The failure of spermiation was also reported in mice lacking the retinoic acid (RA) receptor-alpha (RARA) in SC (Rara(Ser-/-) mutants) and represents, in addition, a feature of vitamin A deficiency that can be readily induced in mice lacking the lecithin:retinol acyltransferase (Lrat(-/-) mutants). Altogether, these findings support the conclusion that RXRB heterodimerized with a RA-liganded RARA transduces signals required in SC for spermatid release.


Assuntos
Colesterol/metabolismo , Regulação da Expressão Gênica , Receptor X Retinoide beta/metabolismo , Células de Sertoli/metabolismo , Espermátides/fisiologia , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Dimerização , Expressão Gênica , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Receptor X Retinoide beta/genética , Receptores Depuradores Classe B/genética
6.
Invest Ophthalmol Vis Sci ; 48(12): 5377-87, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18055784

RESUMO

PURPOSE: To generate a mouse model in which the Lrat gene is selectively disrupted in the retinal pigment epithelium (RPE). To evaluate the effects on the synthesis of retinyl esters and on the expression of other proteins involved in the continuation of the visual cycle. METHODS: A mouse line in which part of the first exon of the Lrat gene has been flanked by loxP sites, was generated and used in the study (Lrat(L3/L3) mice). Heterozygous mice (Lrat(+/L3)) were crossed with mice expressing Cre-recombinase under control of the tyrosinase-related protein-1 (Tyrp1) promoter, which is active selectively in melanin-synthesizing cells such as RPE cells. Accordingly, mice obtained from these crosses should display an RPE-specific disruption of the Lrat gene (Lrat(rpe-/-)). In addition, by crossing CMV-Cre transgenic mice with Lrat(L3/L3) animals, a germline null Lrat knockout (Lrat(L-/L-) mice) was generated. RNA and protein expression, endogenous retinoid levels, and electroretinogram (ERG) analyses were performed on Lrat(rpe-/-) and Lrat(L-)/(L-) mice, to determine the effects of Lrat disruption. Retinoid levels in nonocular tissues were also analyzed for comparison. RESULTS: Analysis of RPE tissues from Lrat(rpe-/-) mice showed absence of Lrat message, lack of Lrat protein expression and consequently a reduced light response in ERG recordings. In addition, RPE cells from Lrat(rpe-/-) showed a strong reduction in their ability to synthesize all-trans retinyl esters, whereas Lrat activity in other tissues known to process retinol was comparable to control Lrat(L3/L3) animals. The Lrat(L-/L-) mice showed no detectable Lrat message, lack of protein expression, and barely detectable ester formation in RPE cells or several other relevant tissues analyzed. CONCLUSIONS: Three Lrat mouse lines with genetic modifications were generated. The Lrat(L-)/(L-) mice displayed features similar to equivalent models previously reported by others. The second mouse line (Lrat(rpe-/-)) displayed loss of Lrat function only in the RPE. The third line possesses functional Lrat in all tissues, but part of the Lrat coding gene was flanked by loxP sites (Lrat(L3/L3)). This feature allows the disruption of this gene in any tissue of choice, by intercrossing with mice in which Cre-recombinase expression is driven by an appropriate tissue-specific promoter.


Assuntos
Aciltransferases/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Epitélio Pigmentado Ocular/enzimologia , Retina/metabolismo , Retinoides/metabolismo , Aciltransferases/biossíntese , Animais , Western Blotting , Eletrorretinografia , Técnica Indireta de Fluorescência para Anticorpo , Vetores Genéticos , Genótipo , Integrases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Mol Cell Biol ; 22(16): 5923-37, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12138202

RESUMO

Human TIF2 (hTIF2) is a member of the p160 family of nuclear receptor coactivators, which includes SRC-1 and p/CIP. Although the functions of hTIF2 and of its mouse homolog (GRIP1 or mTIF2) have been clearly established in vitro, their physiological role remains elusive. Here, we have generated mice lacking mTIF2/GRIP1 and examined their phenotype with a particular emphasis on reproductive functions. TIF2(-/-) mice are viable, but the fertility of both sexes is impaired. Male hypofertility is due to defects in both spermiogenesis (teratozoospermia) and age-dependent testicular degeneration, and TIF2 expression appears to be essential for adhesion of Sertoli cells to germ cells. Female hypofertility is due to a placental hypoplasia that most probably reflects a requirement for maternal TIF2 in decidua stromal cells that face the developing placenta. We conclude that TIF2 plays a critical role in mouse reproductive functions, whereas previous reports have not revealed serious fertility impairment in SRC-1(-/-) or p/CIP(-/-) mutants. Thus, even though the three p160 coactivators exhibit strong sequence homology and similar activity in assays in vitro, they play distinct physiological roles in vivo, as their genetic eliminations result in distinct pathologies.


Assuntos
Fertilidade/fisiologia , Reprodução/fisiologia , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Fatores Etários , Animais , Apoptose/fisiologia , Embrião de Mamíferos/anatomia & histologia , Epididimo/citologia , Epididimo/metabolismo , Feminino , Marcação de Genes , Histona Acetiltransferases , Humanos , Marcação In Situ das Extremidades Cortadas , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Coativador 1 de Receptor Nuclear , Coativador 2 de Receptor Nuclear , Coativador 3 de Receptor Nuclear , Placenta/citologia , Gravidez , Receptores de Esteroides/metabolismo , Testículo/metabolismo , Testículo/ultraestrutura , Transativadores/genética , Fatores de Transcrição/genética
8.
Endocrinology ; 147(1): 96-110, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16210368

RESUMO

As a first step in investigating the role of retinoic acid (RA) in mouse testis, we analyzed the distribution pattern of the enzymes involved in vitamin A storage (lecithin:retinol acyltransferase), RA synthesis (beta-carotene 15,15'-monoxygenase and retinaldehyde dehydrogenases) and RA degradation (cytochrome P450 hydroxylases) as well as those of all isotypes of receptors transducing the RA signal [RA receptors (RARs) and rexinoid receptors (RXRs)]. Our data indicate that in adult testis 1) cytochrome P450 hydroxylase enzymes may generate in peritubular myoid cells a catabolic barrier that prevents circulating RA and RA synthesized by Leydig cells to enter the seminiferous epithelium; 2) the compartmentalization of RA synthesis within this epithelium may modulate, through paracrine mechanisms, the coupling between spermatogonia proliferation and spermatogenesis; 3) retinyl esters synthesized in round spermatids by lecithin:retinol acyltransferase may be transferred and stored in Sertoli cells, in the form of adipose differentiation-related protein-coated lipid droplets. We also show that RARalpha and RXRbeta are confined to Sertoli cells, whereas RARgamma is expressed in spermatogonia and RARbeta, RXRalpha, and RXRgamma are colocalized in step 7-8 spermatids. Correlating these expression patterns with the pathological phenotypes generated in response to RAR and RXR mutations and to postnatal vitamin A deficiency suggests that spermiation requires RXRbeta/RARalpha heterodimers in Sertoli cells, whereas spermatogonia proliferation involves, independently of RXR, two distinct RAR-mediated signaling pathways in both Sertoli cells and spermatogonia. Our data also suggest that the involvement of RA in testis development starts when primary spermatogonia first appear.


Assuntos
Testículo/crescimento & desenvolvimento , Tretinoína/metabolismo , Animais , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/fisiologia , Receptor alfa de Ácido Retinoico , Transdução de Sinais , Testículo/fisiologia
9.
Endocrinology ; 153(1): 438-49, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22045663

RESUMO

Vitamin A is instrumental to mammalian reproduction. Its metabolite, retinoic acid (RA), acts in a hormone-like manner through binding to and activating three nuclear receptor isotypes, RA receptor (RAR)α (RARA), RARß, and RARγ (RARG). Here, we show that 1) RARG is expressed by A aligned (A(al)) spermatogonia, as well as during the transition from A(al) to A(1) spermatogonia, which is known to require RA; and 2) ablation of Rarg, either in the whole mouse or specifically in spermatogonia, does not affect meiosis and spermiogenesis but impairs the A(al) to A(1) transition in the course of some of the seminiferous epithelium cycles. Upon ageing, this phenomenon yields seminiferous tubules containing only spermatogonia and Sertoli cells. Altogether, our findings indicate that RARG cell-autonomously transduces, in undifferentiated spermatogonia of adult testes, a RA signal critical for spermatogenesis. During the prepubertal spermatogenic wave, the loss of RARG function can however be compensated by RARA, as indicated by the normal timing of appearance of meiotic cells in Rarg-null testes. Accordingly, RARG- and RARA-selective agonists are both able to stimulate Stra8 expression in wild-type prepubertal testes. Interestingly, inactivation of Rarg does not impair expression of the spermatogonia differentiation markers Kit and Stra8, contrary to vitamin A deficiency. This latter observation supports the notion that the RA-signaling pathway previously shown to operate in Sertoli cells also participates in spermatogonia differentiation.


Assuntos
Receptores do Ácido Retinoico/metabolismo , Espermatogênese/fisiologia , Espermatogônias/citologia , Espermatogônias/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Sequência de Bases , Primers do DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Meiose , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptores do Ácido Retinoico/deficiência , Receptores do Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Espermatogênese/genética , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Testículo/patologia , Deficiência de Vitamina A/metabolismo , Deficiência de Vitamina A/patologia , Receptor gama de Ácido Retinoico
10.
Environ Health Perspect ; 119(11): 1590-5, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21807577

RESUMO

BACKGROUND: Palate development depends on complex events and is very sensitive to disruption. Accordingly, clefts are the most common congenital malformations worldwide, and a connection is proposed with fetal exposure to toxic factors or environmental contaminants, such as dioxins. There is increasing evidence that dioxin interferes with all-trans-retinoic acid (atRA), a hormone-like signal derived from vitamin A, which plays an essential role during embryonic development. Although similarities have been described between dioxin-induced toxicity and the outcome of altered atRA signaling during palate development, their relationship needs to be clarified. OBJECTIVES: We used a genetic approach to understand the interaction between atRA and dioxin and to identify the cell type targeted by dioxin toxicity during secondary palate formation in mice. METHODS: We analyzed the phenotype of mouse embryos harboring an atRA-sensitive reporter transgene or bearing null mutations for atRA-synthesizing enzymes (RALDH) or atRA receptors (RAR) and maternally exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) at gestation day 10.5. RESULTS: We found that an intact atRA signal was required to enable TCDD to induce cleft palate. This mandatory atRA signal was generated through the activity of RALDH3 in the nasal epithelium and was transduced by RARγ (RARG) in the nasal mesenchyme, where it notably controlled aryl hydrocarbon receptor (Ahr) transcript levels. TCDD also did not alter the developmental pattern of atRA signaling during palate formation. CONCLUSIONS: TCDD-induced alteration of secondary palate development in the mouse appears to depend on atRA signaling, which controls AHR expression. This mechanism is likely conserved throughout vertebrate evolution and may therefore be relevant in humans.


Assuntos
Fissura Palatina/induzido quimicamente , Mesoderma/efeitos dos fármacos , Palato/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/metabolismo , Tretinoína/metabolismo , Animais , Fissura Palatina/etiologia , Fissura Palatina/genética , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Animais , Palato/embriologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores do Ácido Retinoico/metabolismo , Retinal Desidrogenase/metabolismo , Receptor alfa de Ácido Retinoico , Receptor gama de Ácido Retinoico
11.
J Cell Sci ; 121(Pt 19): 3233-42, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18799790

RESUMO

We analysed the phenotypic outcome of a Stra8-null mutation on male meiosis. Because the mutant spermatocytes (1) underwent premeiotic DNA replication, (2) displayed cytological features attesting initiation of recombination and of axial-element assembly, and (3) expressed Spo11 and numerous other meiotic genes, it was concluded that STRA8 is dispensable for meiotic initiation. The few mutant spermatocytes that progressed beyond leptonema showed a prolonged bouquet-stage configuration, asynapsis and heterosynapsis, suggesting function(s) of STRA8 in chromosome pairing. Most importantly, a large number of mutant leptotene spermatocytes underwent premature chromosome condensation, within 24 hours following the meiotic S phase. This phenomenon yielded aberrant metaphase-like cells with 40 univalent chromosomes, similar to normal mitotic metaphases. From these latter observations and from the wild-type pattern of Stra8 expression, we propose that, in preleptotene spermatocytes, STRA8 is involved in the process that leads to stable commitment to the meiotic cell cycle.


Assuntos
Pareamento Cromossômico , Cromossomos de Mamíferos/metabolismo , Meiose , Proteínas/metabolismo , Espermatócitos/citologia , Espermatócitos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Alelos , Animais , Marcação de Genes , Masculino , Prófase Meiótica I , Metáfase , Camundongos , Mutação/genética , Proteínas/genética , Fase S , Espermatogênese , Complexo Sinaptonêmico/metabolismo , Testículo/citologia , Testículo/metabolismo
12.
EMBO J ; 25(24): 5816-25, 2006 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-17124491

RESUMO

Sertoli cells (SC) are instrumental to stem spermatogonia differentiation, a process that critically depends on retinoic acid (RA). We show here that selective ablation of RA receptor alpha (RARalpha) gene in mouse SC, singly (Rara(Ser-/-) mutation) or in combination with RARbeta and RARgamma genes (Rara/b/g(Ser-/-) mutation), abolishes cyclical gene expression in these cells. It additionally induces testis degeneration and delays spermatogonial expression of Stra8, two hallmarks of RA deficiency. As identical defects are generated upon inactivation of RARalpha in the whole organism, our data demonstrate that all the functions exerted by RARalpha in male reproduction are Sertoli cell-autonomous. They further indicate that RARalpha is a master regulator of the cyclical activity of SC and controls paracrine pathways required for spermatogonia differentiation and germ cell survival. Most importantly, we show that the ablation of all RXR (alpha, beta and gamma isotypes) in SC does not recapitulate the phenotype generated upon ablation of all three RARs, thereby providing the first evidence that RARs exert functions in vivo independently of RXRs.


Assuntos
Receptores do Ácido Retinoico/metabolismo , Receptores X de Retinoides/metabolismo , Células de Sertoli/metabolismo , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Envelhecimento , Animais , Contagem de Células , Ciclo Celular , Polaridade Celular , Epitélio , Expressão Gênica , Masculino , Camundongos , Fenótipo , Receptores do Ácido Retinoico/deficiência , Receptor alfa de Ácido Retinoico , Receptor X Retinoide alfa/metabolismo , Receptor X Retinoide beta/metabolismo , Receptor X Retinoide gama/metabolismo , Túbulos Seminíferos/citologia , Serina/genética , Espermatogênese/fisiologia , Testículo/citologia , Testículo/patologia , Receptor gama de Ácido Retinoico
13.
Dev Dyn ; 235(6): 1608-22, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16586441

RESUMO

Using Rbp4-null mice as models, we have established for the first time the kinetics of the spermatogenetic alterations during vitamin A deficiency (VAD). Our data demonstrate that the VAD-induced testicular degeneration arises through the normal maturation of germ cells in a context of spermatogonia differentiation arrest. They indicate that retinoic acid (RA) appears dispensable for the transition of premeiotic to meiotic spermatocytes, meiosis, and spermiogenesis. They confirm that RA plays critical roles in controlling spermatogonia differentiation, spermatid adhesion to Sertoli cells, and spermiation, and suggest that the VAD-induced arrest of spermatogonia differentiation results from simultaneous blocks in RA-dependent events mediated by RA receptor gamma (RARgamma) in spermatogonia and by RARalpha in Sertoli cells. They also provide evidence that expression of major RA-metabolizing enzymes is increased in mouse Sertoli cells upon VAD and that vitamin A-deficient A spermatogonia differ from their RA-sufficient counterparts by the expression of the Stra8 gene.


Assuntos
Retinoides/fisiologia , Proteínas de Ligação ao Retinol/deficiência , Proteínas de Ligação ao Retinol/genética , Espermatogênese/fisiologia , Animais , Marcação de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Plasmáticas de Ligação ao Retinol , Vitamina A/fisiologia
14.
Dev Dyn ; 233(1): 167-76, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15765518

RESUMO

Within cells, retinol (ROL) is bound to cytoplasmic proteins (cellular retinol-binding proteins [CRBPs]), whose proposed function is to protect it from unspecific enzymes through channeling to retinoid-metabolizing pathways. We show that, during development, ROL and retinyl ester levels are decreased in CRBP type 1 (CRBP1) -deficient embryos and fetuses by 50% and 80%, respectively. The steady state level of retinoic acid (RA) is also decreased but to a lesser extent. However, CRBP1-null fetuses do not exhibit the abnormalities characteristic of a vitamin A-deficiency syndrome. Neither CRBP1 deficiency alters the expression patterns of RA-responding genes during development, nor does CRBP1 availability modify the expression of an RA-dependent gene in primary embryonic fibroblasts treated with ROL. Therefore, CRBP1 is required in prenatal life to maintain normal amounts of ROL and to ensure its efficient storage but seems of secondary importance for RA synthesis, at least under conditions of maternal vitamin A sufficiency.


Assuntos
Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Ligação ao Retinol/metabolismo , Vitamina A/metabolismo , Animais , Fibroblastos/metabolismo , Genes Reporter , Homeostase/fisiologia , Camundongos , Proteínas de Ligação ao Retinol/genética , Proteínas Celulares de Ligação ao Retinol , Tretinoína/metabolismo
15.
Development ; 132(21): 4789-800, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16207763

RESUMO

Using genetic approaches in the mouse, we show that the primary target tissue of retinoic acid (RA) action during eye morphogenesis is not the retina nor the corneal ectoderm, which both express RA-synthesizing retinaldehyde dehydrogenases (RALDH1 and RALDH3), but the neural crest cell-derived periocular mesenchyme (POM), which is devoid of RALDH. In POM, the effects of the paracrine RA signal are mediated by the nuclear RA receptors heterodimers RXRalpha/RARbeta and RXRalpha/RARgamma. These heterodimers appear to control: (1) the remodeling of the POM through activation of Eya2-related apoptosis; (2) the expression of Foxc1 and Pitx2, which play crucial roles in anterior eye segment development; and (3) the growth of the ventral retina. We additionally show that RALDH1 and RALDH3 are the only enzymes that are required for RA synthesis in the eye region from E10.5 to E13.5, and that patterning of the dorsoventral axis of the retina does not require RA.


Assuntos
Desenvolvimento Embrionário , Olho/embriologia , Morfogênese/efeitos dos fármacos , Crista Neural/citologia , Tretinoína/farmacologia , Aldeído Oxirredutases/fisiologia , Animais , Olho/crescimento & desenvolvimento , Mesoderma , Camundongos , Camundongos Transgênicos , Comunicação Parácrina , Receptores do Ácido Retinoico/fisiologia
16.
Dev Dyn ; 226(1): 103-6, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12508230

RESUMO

Ovaries from adult mice lacking both estrogen receptors ERalpha and ERbeta (ERalphabetaKO mice) contain abnormal cells sharing morphologic features with Sertoli cells, which are located mainly in the interstitial compartment. We show here that these cells express the Sertoli cell markers TIF1beta, TIF2, and Sox9. In ERalphabetaKO ovaries, Sox9 is expressed by granulosa cells before the morphologic appearance of Sertoli cells, but neither by granulosa cell precursors nor by non-Sertolian interstitial cells. These findings suggest that functional Sertoli cells can transdifferentiate from mature granulosa cells devoid of estrogen receptors as a result of Sox9 expression.


Assuntos
Células da Granulosa/metabolismo , Proteínas de Grupo de Alta Mobilidade/biossíntese , Ovário/metabolismo , Receptores de Estrogênio/genética , Fatores de Transcrição/biossíntese , Animais , Diferenciação Celular , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Mutação , Fatores de Transcrição SOX9 , Células de Sertoli/metabolismo , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA