Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chromosoma ; 127(3): 341-359, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29582139

RESUMO

During mammalian meiotic prophase, homologous chromosomes connect through the formation of the synaptonemal complex (SC). SYCP3 is a component of the lateral elements of the SC. We have generated transgenic mice expressing N- or C-terminal fluorescent-tagged SYCP3 (mCherry-SYCP3 (CSYCP) and SYCP3-mCherry (SYCPC)) to study SC dynamics and chromosome movements in vivo. Neither transgene rescued meiotic aberrations in Sycp3 knockouts, but CSYCP could form short axial element-like structures in the absence of endogenous SYCP3. On the wild-type background, both fusion proteins localized to the axes of the SC together with endogenous SYCP3, albeit with delayed initiation (from pachytene) in spermatocytes. Around 40% of CSYCP and SYCPC that accumulated on the SC was rapidly exchanging with other tagged proteins, as analyzed by fluorescent recovery after photobleaching (FRAP) assay. We used the CSYCP transgenic mice for further live cell analyses and observed synchronized bouquet configurations in living cysts of two or three zygotene oocyte nuclei expressing CSYCP, which presented cycles of telomere clustering and dissolution. Rapid chromosome movements were observed in both zygotene oocytes and pachytene spermatocytes, but rotational movements of the nucleus were more clear in oocytes. In diplotene spermatocytes, desynapsis was found to proceed in a discontinuous manner, whereby even brief chromosome re-association events were observed. Thus, this live imaging approach can be used to follow changes in the dynamic behavior of the nucleus and chromatin, in normal mice and different infertile mouse models.


Assuntos
Cromossomos de Mamíferos , Ovário/metabolismo , Túbulos Seminíferos/metabolismo , Complexo Sinaptonêmico/genética , Animais , Biomarcadores , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Feminino , Expressão Gênica , Técnicas de Inativação de Genes , Masculino , Meiose/genética , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/genética , Oócitos/metabolismo , Fenótipo , Espermatócitos/metabolismo , Testículo , Transgenes
2.
BMC Genomics ; 16: 291, 2015 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-25884295

RESUMO

BACKGROUND: In mammalian meiotic prophase, homologous chromosome recognition is aided by formation and repair of programmed DNA double-strand breaks (DSBs). Subsequently, stable associations form through homologous chromosome synapsis. In male mouse meiosis, the largely heterologous X and Y chromosomes synapse only in their short pseudoautosomal regions (PARs), and DSBs persist along the unsynapsed non-homologous arms of these sex chromosomes. Asynapsis of these arms and the persistent DSBs then trigger transcriptional silencing through meiotic sex chromosome inactivation (MSCI), resulting in formation of the XY body. This inactive state is partially maintained in post-meiotic haploid spermatids (postmeiotic sex chromatin repression, PSCR). For the human, establishment of MSCI and PSCR have also been reported, but X-linked gene silencing appears to be more variable compared to mouse. To gain more insight into the regulation and significance of MSCI and PSCR among different eutherian species, we have performed a global analysis of XY pairing dynamics, DSB repair, MSCI and PSCR in the domestic dog (Canis lupus familiaris), for which the complete genome sequence has recently become available, allowing a thorough comparative analyses. RESULTS: In addition to PAR synapsis between X and Y, we observed extensive self-synapsis of part of the dog X chromosome, and rapid loss of known markers of DSB repair from that part of the X. Sequencing of RNA from purified spermatocytes and spermatids revealed establishment of MSCI. However, the self-synapsing region of the X displayed higher X-linked gene expression compared to the unsynapsed area in spermatocytes, and was post-meiotically reactivated in spermatids. In contrast, genes in the PAR, which are expected to escape MSCI, were expressed at very low levels in both spermatocytes and spermatids. Our comparative analysis was then used to identify two X-linked genes that may escape MSCI in spermatocytes, and 21 that are specifically re-activated in spermatids of human, mouse and dog. CONCLUSIONS: Our data indicate that MSCI is incomplete in the dog. This may be partially explained by extensive, but transient, self-synapsis of the X chromosome, in association with rapid completion of meiotic DSB repair. In addition, our comparative analysis identifies novel candidate male fertility genes.


Assuntos
Cromossomos de Mamíferos/metabolismo , Cães/genética , Meiose , Cromossomos Sexuais/metabolismo , Espermatogênese , Inativação do Cromossomo X , Animais , Animais Domésticos , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Cães/metabolismo , Humanos , Masculino , Camundongos , Espermatócitos/citologia , Espermatócitos/metabolismo , Testículo
3.
BMC Genomics ; 11: 367, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20537150

RESUMO

BACKGROUND: The ubiquitin-conjugating enzyme HR6B is required for spermatogenesis in mouse. Loss of HR6B results in aberrant histone modification patterns on the trancriptionally silenced X and Y chromosomes (XY body) and on centromeric chromatin in meiotic prophase. We studied the relationship between these chromatin modifications and their effects on global gene expression patterns, in spermatocytes and spermatids. RESULTS: HR6B is enriched on the XY body and on centromeric regions in pachytene spermatocytes. Global gene expression analyses revealed that spermatid-specific single- and multicopy X-linked genes are prematurely expressed in Hr6b knockout spermatocytes. Very few other differences in gene expression were observed in these cells, except for upregulation of major satellite repeat transcription. In contrast, in Hr6b knockout spermatids, 7298 genes were differentially expressed; 65% of these genes was downregulated, but we observed a global upregulation of gene transcription from the X chromosome. In wild type spermatids, approximately 20% of the single-copy X-linked genes reach an average expression level that is similar to the average expression from autosomes. CONCLUSIONS: Spermatids maintain an enrichment of repressive chromatin marks on the X chromosome, originating from meiotic prophase, but this does not interfere with transcription of the single-copy X-linked genes that are reactivated or specifically activated in spermatids. HR6B represses major satellite repeat transcription in spermatocytes, and functions in the maintenance of X chromosome silencing in spermatocytes and spermatids. It is discussed that these functions involve modification of chromatin structure, possibly including H2B ubiquitylation.


Assuntos
Espermátides/metabolismo , Espermatócitos/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Inativação do Cromossomo X , Cromossomo X/genética , Animais , Proteínas de Ciclo Celular/genética , Centrômero/genética , Centrômero/metabolismo , Cromatina/genética , Cromatina/metabolismo , Dosagem de Genes/genética , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Genes Ligados ao Cromossomo X/genética , Histonas/genética , Histonas/metabolismo , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Especificidade de Órgãos , Fosfoproteínas/genética , Testículo/metabolismo , Transcrição Gênica , Ativação Transcricional , Enzimas de Conjugação de Ubiquitina/deficiência , Enzimas de Conjugação de Ubiquitina/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Regulação para Cima , Cromossomo X/metabolismo , Cromossomo Y/genética
4.
J Cell Sci ; 123(Pt 3): 331-9, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20053632

RESUMO

The cytoplasmic chromatoid body (CB) organizes mRNA metabolism and small regulatory RNA pathways, in relation to haploid gene expression, in mammalian round spermatids. However, little is known about functions and fate of the CB at later steps of spermatogenesis, when elongating spermatids undergo chromatin compaction and transcriptional silencing. In mouse elongating spermatids, we detected accumulation of the testis-specific serine/threonine kinases TSSK1 and TSSK2, and the substrate TSKS, in a ring-shaped structure around the base of the flagellum and in a cytoplasmic satellite, both corresponding to structures described to originate from the CB. At later steps of spermatid differentiation, the ring is found at the caudal end of the newly formed mitochondrial sheath. Targeted deletion of the tandemly arranged genes Tssk1 and Tssk2 in mouse resulted in male infertility, with loss of the CB-derived ring structure, and with elongating spermatids possessing a collapsed mitochondrial sheath. These results reveal TSSK1- and TSSK2-dependent functions of a transformed CB in post-meiotic cytodifferentiation of spermatids.


Assuntos
Grânulos Citoplasmáticos/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Espermátides/enzimologia , Espermátides/metabolismo , Testículo/enzimologia , Animais , Western Blotting , Proteínas do Citoesqueleto , Eletroforese em Gel de Poliacrilamida , Feminino , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Fosfoproteínas , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Espermátides/ultraestrutura , Espermatogênese/genética , Espermatogênese/fisiologia , Testículo/ultraestrutura
5.
PLoS Genet ; 5(5): e1000466, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19461881

RESUMO

During meiotic prophase in male mammals, the heterologous X and Y chromosomes remain largely unsynapsed, and meiotic sex chromosome inactivation (MSCI) leads to formation of the transcriptionally silenced XY body. In birds, the heterogametic sex is female, carrying Z and W chromosomes (ZW), whereas males have the homogametic ZZ constitution. During chicken oogenesis, the heterologous ZW pair reaches a state of complete heterologous synapsis, and this might enable maintenance of transcription of Z- and W chromosomal genes during meiotic prophase. Herein, we show that the ZW pair is transiently silenced, from early pachytene to early diplotene using immunocytochemistry and gene expression analyses. We propose that ZW inactivation is most likely achieved via spreading of heterochromatin from the W on the Z chromosome. Also, persistent meiotic DNA double-strand breaks (DSBs) may contribute to silencing of Z. Surprisingly, gammaH2AX, a marker of DSBs, and also the earliest histone modification that is associated with XY body formation in mammalian and marsupial spermatocytes, does not cover the ZW during the synapsed stage. However, when the ZW pair starts to desynapse, a second wave of gammaH2AX accumulates on the unsynapsed regions of Z, which also show a reappearance of the DSB repair protein RAD51. This indicates that repair of meiotic DSBs on the heterologous part of Z is postponed until late pachytene/diplotene, possibly to avoid recombination with regions on the heterologously synapsed W chromosome. Two days after entering diplotene, the Z looses gammaH2AX and shows reactivation. This is the first report of meiotic sex chromosome inactivation in a species with female heterogamety, providing evidence that this mechanism is not specific to spermatogenesis. It also indicates the presence of an evolutionary force that drives meiotic sex chromosome inactivation independent of the final achievement of synapsis.


Assuntos
Galinhas/genética , Meiose/genética , Cromossomos Sexuais/genética , Acetilação , Animais , Sequência de Bases , Galinhas/metabolismo , Quebras de DNA de Cadeia Dupla , Metilação de DNA , Primers do DNA/genética , Reparo do DNA , Mecanismo Genético de Compensação de Dose , Feminino , Perfilação da Expressão Gênica , Inativação Gênica , Histonas/química , Histonas/metabolismo , Hibridização in Situ Fluorescente , Lisina/química , Masculino , Microscopia de Fluorescência , Oócitos/citologia , Oócitos/metabolismo , Oogênese/genética , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Filogenia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cromossomos Sexuais/metabolismo , Espermatogênese/genética , Complexo Sinaptonêmico/genética , Complexo Sinaptonêmico/metabolismo
6.
PLoS One ; 4(5): e5616, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19440388

RESUMO

BACKGROUND: In female mammalian cells, random X chromosome inactivation (XCI) equalizes the dosage of X-encoded gene products to that in male cells. XCI is a stochastic process, in which each X chromosome has a probability to be inactivated. To obtain more insight in the factors setting up this probability, we studied the role of the X to autosome (X ratio A) ratio in initiation of XCI, and have used the experimental data in a computer simulation model to study the cellular population dynamics of XCI. METHODOLOGY/PRINCIPAL FINDINGS: To obtain more insight in the role of the XratioA ratio in initiation of XCI, we generated triploid mouse ES cells by fusion of haploid round spermatids with diploid female and male ES cells. These fusion experiments resulted in only XXY triploid ES cells. XYY and XXX ES lines were absent, suggesting cell death related either to insufficient X-chromosomal gene dosage (XYY) or to inheritance of an epigenetically modified X chromosome (XXX). Analysis of active (Xa) and inactive (Xi) X chromosomes in the obtained triploid XXY lines indicated that the initiation frequency of XCI is low, resulting in a mixed population of XaXiY and XaXaY cells, in which the XaXiY cells have a small proliferative advantage. This result, and findings on XCI in diploid and tetraploid ES cell lines with different X ratio A ratios, provides evidence that the X ratio A ratio determines the probability for a given X chromosome to be inactivated. Furthermore, we found that the kinetics of the XCI process can be simulated using a probability for an X chromosome to be inactivated that is proportional to the X ratio A ratio. These simulation studies re-emphasize our hypothesis that the probability is a function of the concentration of an X-encoded activator of XCI, and of X chromosome specific allelic properties determining the threshold for this activator. CONCLUSIONS: The present findings reveal that the probability for an X chromosome to be inactivated is proportional to the X ratio A ratio. This finding supports the presence of an X-encoded activator of the XCI process.


Assuntos
Alelos , Genes Ligados ao Cromossomo X/genética , Inativação do Cromossomo X/genética , Cromossomo X/genética , Animais , Células Cultivadas , Feminino , Hibridização in Situ Fluorescente , Cariotipagem , Masculino , Camundongos , Poliploidia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Mol Cell Endocrinol ; 292(1-2): 69-78, 2008 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-18656523

RESUMO

A novel mutation F826L located within the ligand binding domain (LBD) of the human androgen receptor (AR) was investigated. This mutation was found in a boy with severe penoscrotal hypospadias (classified as 46,XY DSD). The AR mutant F826L appeared to be indistinguishable from the wild-type AR, with respect to ligand binding affinity, transcriptional activation of MMTV-luciferase and ARE2-TATA-luciferase reporter genes, protein level in genital skin fibroblasts (GSFs), and sub-cellular distribution in transfected cells. However, an at least two-fold higher NH2-/COOH-terminal domain interaction was found in luciferase and GST pull-down assays. A two-fold increase was also observed for TIF2 (transcription intermediary factor 2) co-activation of the AR F826L COOH-terminal domain. This increase could not be explained by a higher stability of the mutant protein, which was within wild-type range. Repression of transactivation by the nuclear receptor co-repressor (N-CoR) was not affected by the AR F826L mutation. The observed properties of AR F826L would be in agreement with an increased activity rather than with a partial defective AR transcriptional activation. It is concluded that the penoscrotal hypospadias in the present case is caused by an as yet unknown mechanism, which still may involve the mutant AR.


Assuntos
Substituição de Aminoácidos , Síndrome de Resistência a Andrógenos/genética , Mutação/genética , Coativador 2 de Receptor Nuclear/metabolismo , Receptores Androgênicos/química , Receptores Androgênicos/genética , Linhagem Celular , Pré-Escolar , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Prepúcio do Pênis/citologia , Humanos , Imunoprecipitação , Lactente , Ligantes , Masculino , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Correpressor 1 de Receptor Nuclear , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Receptores Androgênicos/metabolismo , Proteínas Repressoras/metabolismo , Frações Subcelulares/metabolismo , Ativação Transcricional/genética
8.
J Cell Sci ; 120(Pt 11): 1841-51, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17488778

RESUMO

Mono-ubiquitylated H2A marks the transcriptionally silenced XY body during male meiotic prophase. Concomitant with H2A(K119ub1), the ubiquitin-conjugating enzyme HR6B is also enriched on the XY body. We analyzed H2A and H2B ubiquitylation in Hr6b-knockout mouse spermatocytes, but no global changes were detected. Next, we analyzed phosphorylation of the threonine residues T120 and T119 that are adjacent to the K119 and K120 target sites for ubiquitylation in H2A and H2B, respectively. In wild-type cells, H2A(T120ph) and H2B(T119ph) mark meiotically unpaired and silenced chromatin, including the XY body. In Hr6b-knockout spermatocytes, the H2B(T119ph) signal was unchanged, but H2A(T120ph) was enhanced from late pachytene until metaphase I. Furthermore, we found increased H3(K4) dimethylation on the X and Y chromosomes of diplotene Hr6b-knockout spermatocytes, persisting into postmeiotic round spermatids. In these cells, the X and Y chromosomes maintained an unchanged H3(K9m2) level, even when this modification was lost from centromeric heterochromatin. Analysis of gene expression showed derepression of X chromosome genes in postmeiotic Hr6b-knockout spermatids. We conclude that HR6B exerts control over different histone modifications in spermatocytes and spermatids, and that this function contributes to the postmeiotic maintenance of X chromosome silencing.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Regulação da Expressão Gênica/genética , Histonas/metabolismo , Enzimas de Conjugação de Ubiquitina/deficiência , Enzimas de Conjugação de Ubiquitina/metabolismo , Cromossomo X/genética , Animais , Heterocromatina/metabolismo , Masculino , Meiose , Metáfase , Metilação , Camundongos , Camundongos Knockout , Nucleossomos/metabolismo , Fosforilação , Espermatócitos/citologia , Espermatócitos/metabolismo , Ubiquitina/metabolismo , Cromossomo X/metabolismo
9.
Genes Dev ; 19(20): 2501-15, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16230537

RESUMO

CLIP-170 is a microtubule "plus-end-tracking protein" implicated in the control of microtubule dynamics, dynactin localization, and the linking of endosomes to microtubules. To investigate the function of mouse CLIP-170, we generated CLIP-170 knockout and GFP-CLIP-170 knock-in alleles. Residual CLIP-170 is detected in lungs and embryos of homozygous CLIP-170 knockout mice, but not in other tissues and cell types, indicating that we have generated a hypomorphic mutant. Homozygous CLIP-170 knockout mice are viable and appear normal. However, male knockout mice are subfertile and produce sperm with abnormal heads. Using the knock-in mice, we followed GFP-CLIP-170 expression and behavior in dissected, live testis tubules. We detect plus-end-tracking GFP-CLIP-170 in spermatogonia. As spermatogenesis proceeds, GFP-CLIP-170 expression increases and the fusion protein strongly marks syncytia of differentiated spermatogonia and early prophase spermatocytes. Subsequently GFP-CLIP-170 levels drop, but during spermiogenesis (post-meiotic development), GFP-CLIP-170 accumulates again and is present on spermatid manchettes and centrosomes. Bleaching studies show that, as spermatogenesis progresses, GFP-CLIP-170 converts from a mobile plus-end-tracking protein to a relatively immobile protein. We propose that CLIP-170 has a structural function in the male germline, in particular in spermatid differentiation and sperm head shaping.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Espermátides/metabolismo , Espermatogênese/fisiologia , Animais , Centrossomo/metabolismo , Centrossomo/ultraestrutura , Endossomos/metabolismo , Endossomos/ultraestrutura , Imunofluorescência/métodos , Homozigoto , Masculino , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Proteínas de Neoplasias/genética , Transporte Proteico , Cabeça do Espermatozoide/metabolismo , Cabeça do Espermatozoide/ultraestrutura , Espermátides/ultraestrutura
10.
Endocrinology ; 146(8): 3558-66, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15878962

RESUMO

Follicle development in the mammalian ovary requires interactions among the oocyte, granulosa cells, and theca cells, coordinating gametogenesis and steroidogenesis. Here we show that granulosa cells of growing follicles in mouse ovary act as a source of hedgehog signaling. Expression of Indian hedgehog and desert hedgehog mRNAs initiates in granulosa cells at the primary follicle stage, and we find induced expression of the hedgehog target genes Ptch1 and Gli1, in the surrounding pre-theca cell compartment. Cyclopamine, a highly specific hedgehog signaling antagonist, inhibits this induced expression of target genes in cultured neonatal mouse ovaries. The theca cell compartment remains a target of hedgehog signaling throughout follicle development, showing induced expression of the hedgehog target genes Ptch1, Ptch2, Hip1, and Gli1. In periovulatory follicles, a dynamic synchrony between loss of hedgehog expression and loss of induced target gene expression is observed. Oocytes are unable to respond to hedgehog because they lack expression of the essential signal transducer Smo (smoothened). The present results point to a prominent role of hedgehog signaling in the communication between granulosa cells and developing theca cells.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Ovário/fisiologia , Células Tecais/fisiologia , Transativadores/fisiologia , Animais , Animais Recém-Nascidos , Indução Embrionária , Feminino , Proteínas Hedgehog , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos , Ovário/crescimento & desenvolvimento , Receptores Patched , Receptor Patched-1 , Receptor Patched-2 , RNA Mensageiro/genética , Receptores de Superfície Celular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Superovulação , Transativadores/genética , Transcrição Gênica
11.
Mol Cell Biol ; 25(3): 1041-53, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15657431

RESUMO

During meiotic prophase in male mammals, the X and Y chromosomes are incorporated in the XY body. This heterochromatic body is transcriptionally silenced and marked by increased ubiquitination of histone H2A. This led us to investigate the relationship between histone H2A ubiquitination and chromatin silencing in more detail. First, we found that ubiquitinated H2A also marks the silenced X chromosome of the Barr body in female somatic cells. Next, we studied a possible relationship between H2A ubiquitination, chromatin silencing, and unpaired chromatin in meiotic prophase. The mouse models used carry an unpaired autosomal region in male meiosis or unpaired X and Y chromosomes in female meiosis. We show that ubiquitinated histone H2A is associated with transcriptional silencing of large chromatin regions. This silencing in mammalian meiotic prophase cells concerns unpaired chromatin regions and resembles a phenomenon described for the fungus Neurospora crassa and named meiotic silencing by unpaired DNA.


Assuntos
Inativação Gênica/fisiologia , Histonas/metabolismo , Meiose/genética , Cromatina Sexual/metabolismo , Aberrações dos Cromossomos Sexuais , Animais , Núcleo Celular/genética , Núcleo Celular/metabolismo , Eletroforese em Gel Bidimensional , Feminino , Humanos , Masculino , Meiose/fisiologia , Camundongos , Oócitos/metabolismo , Prófase/fisiologia , Ratos , Espermatócitos/metabolismo
12.
Nucleic Acids Res ; 32(21): 6425-36, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15585666

RESUMO

In mouse spermatogenesis, differentiating germ line cells initiate expression of specific genes at subsequent developmental steps. The Calmegin (Clgn) gene is first expressed in meiotic prophase, in primary spermatocytes, and encodes a protein that acts as a chaperone. To identify testis-specific transcription factors that control expression of the Clgn gene in spermatogenesis, we performed a yeast one-hybrid screening with a Clgn promoter sequence as bait DNA. This screening resulted in the identification of mouse Tcfl5 as a candidate Clgn promoter-binding protein. Tcfl5 is a member of the basic helix-loop-helix (bHLH) family of transcription factors, and mouse Tcfl5 shows 83% amino acid sequence identity with human TCFL5. Gel-shift and yeast one-hybrid experiments showed that Tcfl5 interacts with a non-canonical CACGCG site that is present in the Clgn promoter. By using northern blot, RT-PCR and in situ hybridization, mouse Tcfl5 mRNA was detected only in testis, with the highest expression level in primary spermatocytes and round spermatids. The highest level of Tcfl5 protein was found in primary spermatocytes at the diplotene stage of meiotic prophase, where the protein colocalizes with transcriptionally active chromatin.


Assuntos
Calnexina/genética , Regiões Promotoras Genéticas , Espermatogênese , Testículo/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Sítios de Ligação , Proteínas de Ligação ao Cálcio , Sequências Hélice-Alça-Hélice , Masculino , Camundongos , Chaperonas Moleculares , Dados de Sequência Molecular , Alinhamento de Sequência , Espermatócitos/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética
13.
J Cell Sci ; 117(Pt 21): 5023-33, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15383616

RESUMO

In replicative damage bypass (RDB) in yeast, the ubiquitin-conjugating enzyme RAD6 interacts with the ubiquitin ligase RAD18. In the mouse, these enzymes are represented by two homologs of RAD6, HR6a and HR6b, and one homolog of RAD18, Rad18Sc. Expression of these genes and the encoded proteins is ubiquitous, but there is relatively high expression in the testis. We have studied the subcellular localization by immunostaining Rad18Sc and other RDB proteins in mouse primary spermatocytes passing through meiotic prophase in spermatogenesis. The highest Rad18Sc protein level is found at pachytene and diplotene, and the protein localizes mainly to the XY body, a subnuclear region that contains the transcriptionally inactivated X and Y chromosomes. In spermatocytes that carry translocations for chromosomes 1 and 13, Rad18Sc protein concentrates on translocation bivalents that are not fully synapsed. The partly synapsed bivalents are often localized in the vicinity of the XY body, and show a very low level of RNA polymerase II, indicating that the chromatin is in a silent configuration similar to transcriptional silencing of the XY body. Thus, Rad18Sc localizes to unsynapsed and silenced chromosome segments during the male meiotic prophase. All known functions of RAD18 in yeast are related to RDB. However, in contrast to Rad18Sc, expression of UBC13 and poleta, known to be involved in subsequent steps of RDB, appears to be diminished in the XY body and regions containing the unpaired translocation bivalents. Taken together, these observations suggest that the observed subnuclear localization of Rad18Sc may involve a function outside the context of RDB. This function is probably related to a mechanism that signals the presence of unsynapsed chromosomal regions and subsequently leads to transcriptional silencing of these regions during male meiotic prophase.


Assuntos
Cromossomos/ultraestrutura , Proteínas de Ligação a DNA/biossíntese , Meiose , Cromossomo X , Cromossomo Y , Animais , Núcleo Celular/metabolismo , Cromatina/metabolismo , Reparo do DNA , DNA Polimerase Dirigida por DNA/biossíntese , Regulação da Expressão Gênica , Inativação Gênica , Heterozigoto , Immunoblotting , Masculino , Camundongos , Modelos Genéticos , Prófase , Ligação Proteica , Espermatócitos/metabolismo , Testículo/metabolismo , Fatores de Tempo , Transcrição Gênica , Translocação Genética , Enzimas de Conjugação de Ubiquitina/biossíntese
14.
Mol Cell Biol ; 24(12): 5485-95, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15169909

RESUMO

The Saccharomyces cerevisiae RAD6 protein is required for a surprising diversity of cellular processes, including sporulation and replicational damage bypass of DNA lesions. In mammals, two RAD6-related genes, HR6A and HR6B, encode highly homologous proteins. Here, we describe the phenotype of cells and mice deficient for the mHR6A gene. Just like mHR6B knockout mouse embryonic fibroblasts, mHR6A-deficient cells appear to have normal DNA damage resistance properties, but mHR6A knockout male and female mice display a small decrease in body weight. The necessity for at least one functional mHR6A (X-chromosomal) or mHR6B (autosomal) allele in all somatic cell types is supported by the fact that neither animals lacking both proteins nor females with only one intact mHR6A allele are viable. In striking contrast to mHR6B knockout males, which show a severe spermatogenic defect, mHR6A knockout males are normally fertile. However, mHR6A knockout females fail to produce offspring despite a normal ovarian histology and ovulation. The absence of mHR6A in oocytes prevents development beyond the embryonic two-cell stage but does not result in an aberrant methylation pattern of histone H3 at this early stage of mouse embryonic development. These observations support redundant but dose-dependent roles for HR6A and HR6B in somatic cell types and germ line cells in mammals.


Assuntos
Reparo do DNA , Desenvolvimento Embrionário e Fetal/fisiologia , Enzimas de Conjugação de Ubiquitina/fisiologia , Alelos , Animais , Sequência de Bases , DNA Complementar/genética , Desenvolvimento Embrionário e Fetal/genética , Feminino , Fertilidade , Marcação de Genes , Histonas/química , Histonas/metabolismo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/metabolismo , Fenótipo , Gravidez , Enzimas de Conjugação de Ubiquitina/deficiência , Enzimas de Conjugação de Ubiquitina/genética
15.
Reprod Biol Endocrinol ; 1: 3, 2003 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-12646048

RESUMO

Several in vivo studies have reported the presence of immunoreactive transforming growth factor-beta's (TGF-beta's) in testicular cells at defined stages of their differentiation. The most pronounced changes in TGF-beta1 and TGF-beta2 immunoreactivity occurred during spermatogenesis. In the present study we have investigated whether germ cells and Sertoli cells are able to secrete bioactive TGF-beta's in vitro, using the CCl64 mink lung epithelial cell line as bioassay for the measurement of TGF-beta. In cellular lysates, TGF-beta bioactivity was only observed following heat-treatment, indicating that within these cells TGF-beta is present in a latent form. To our surprise, active TGF-beta could be detected in the culture supernatant of germ cells and Sertoli cells without prior heat-treatment. This suggests that these cells not only produce and release TGF-beta in a latent form, but that they also release a factor which can convert latent TGF-beta into its active form. Following heat-activation of these culture supernatant's, total TGF-beta bioactivity increased 6- to 9-fold. Spermatocytes are the cell type that releases most bioactive TGF-beta during a 24 h culture period, although round and elongated spermatids and Sertoli cells also secrete significant amounts of TGF-beta. The biological activity of TGF-beta could be inhibited by neutralizing antibodies against TGF-beta1 (spermatocytes and round spermatids) and TGF-beta2 (round and elongating spermatids). TGF-beta activity in the Sertoli cell culture supernatant was inhibited slightly by either the TGF-beta1 and TGF-beta2 neutralizing antibody. These in vitro data suggest that germ cells and Sertoli cells release latent TGF-beta's. Following secretion, the TGF-beta's are converted to a biological active form that can interact with specific TGF-beta receptors. These results strengthen the hypothesis that TGF-beta's may play a physiological role in germ cell proliferation/differentiation and Sertoli cell function.


Assuntos
Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Bioensaio , Diferenciação Celular , Divisão Celular , Linhagem Celular , Células Cultivadas/metabolismo , Meios de Cultivo Condicionados/farmacologia , Pulmão , Masculino , Vison , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Ratos , Ratos Wistar , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Espermátides/metabolismo , Espermatócitos/metabolismo , Fator de Crescimento Transformador beta1 , Fator de Crescimento Transformador beta2
16.
Mol Cell Biol ; 23(4): 1151-62, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12556476

RESUMO

The ubiquitin-conjugating enzymes HR6A and HR6B are the two mammalian homologs of Saccharomyces cerevisiae RAD6. In yeast, RAD6 plays an important role in postreplication DNA repair and in sporulation. HR6B knockout mice are viable, but spermatogenesis is markedly affected during postmeiotic steps, leading to male infertility. In the present study, increased apoptosis of HR6B knockout primary spermatocytes was detected during the first wave of spermatogenesis, indicating that HR6B performs a primary role during the meiotic prophase. Detailed analysis of HR6B knockout pachytene nuclei showed major changes in the synaptonemal complexes. These complexes were found to be longer. In addition, we often found depletion of synaptonemal complex proteins from near telomeric regions in the HR6B knockout pachytene nuclei. Finally, we detected an increased number of foci containing the mismatch DNA repair protein MLH1 in these nuclei, reflecting a remarkable and consistent increase (20 to 25%) in crossing-over frequency. The present findings reveal a specific requirement for the ubiquitin-conjugating activity of HR6B in relation to dynamic aspects of the synaptonemal complex and meiotic recombination in spermatocytes.


Assuntos
Troca Genética , Ligases/metabolismo , Meiose , Prófase/genética , Complexo Sinaptonêmico/ultraestrutura , Enzimas de Conjugação de Ubiquitina , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte , Morte Celular/genética , Reparo do DNA/genética , Ligases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 1 Homóloga a MutL , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Espermatócitos/fisiologia , Complexo Sinaptonêmico/genética , Telômero/genética , Telômero/metabolismo , Testículo/citologia , Testículo/fisiologia , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...