Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell Rep ; 25(7): 1912-1923.e9, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30428357

RESUMO

We and others have recently reported that the SMC protein Smchd1 is a regulator of chromosome conformation. Smchd1 is critical for the structure of the inactive X chromosome and at autosomal targets such as the Hox genes. However, it is unknown how Smchd1 is recruited to these sites. Here, we report that Smchd1 localizes to the inactive X via the Xist-HnrnpK-PRC1 (polycomb repressive complex 1) pathway. Contrary to previous reports, Smchd1 does not bind Xist or other RNA molecules with any specificity. Rather, the localization of Smchd1 to the inactive X is H2AK119ub dependent. Following perturbation of this interaction, Smchd1 is destabilized, which has consequences for gene silencing genome-wide. Our work adds Smchd1 to the PRC1 silencing pathway for X chromosome inactivation.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Complexo Repressor Polycomb 1/metabolismo , RNA Longo não Codificante/metabolismo , Inativação do Cromossomo X/genética , Animais , Sequência de Bases , Diferenciação Celular , Feminino , Genoma , Histonas/metabolismo , Lisina/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Oligonucleotídeos/metabolismo , Transporte Proteico
2.
Nat Struct Mol Biol ; 25(9): 766-777, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30127357

RESUMO

The regulation of higher-order chromatin structure is complex and dynamic, and a full understanding of the suite of mechanisms governing this architecture is lacking. Here, we reveal the noncanonical SMC protein Smchd1 to be a novel regulator of long-range chromatin interactions in mice, and we add Smchd1 to the canon of epigenetic proteins required for Hox-gene regulation. The effect of losing Smchd1-dependent chromatin interactions has varying outcomes that depend on chromatin context. At autosomal targets transcriptionally sensitive to Smchd1 deletion, we found increased short-range interactions and ectopic enhancer activation. In contrast, the inactive X chromosome was transcriptionally refractive to Smchd1 ablation, despite chromosome-wide increases in short-range interactions. In the inactive X, we observed spreading of trimethylated histone H3 K27 (H3K27me3) domains into regions not normally decorated by this mark. Together, these data suggest that Smchd1 is able to insulate chromatin, thereby limiting access to other chromatin-modifying proteins.


Assuntos
Cromatina/metabolismo , Proteínas Cromossômicas não Histona/fisiologia , Genes Homeobox , Família Multigênica , Cromossomo X , Animais , Proteínas Cromossômicas não Histona/genética , Elementos Facilitadores Genéticos , Deleção de Genes , Inativação Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Proc Natl Acad Sci U S A ; 112(27): E3535-44, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26091879

RESUMO

Structural maintenance of chromosomes flexible hinge domain containing 1 (Smchd1) is an epigenetic repressor with described roles in X inactivation and genomic imprinting, but Smchd1 is also critically involved in the pathogenesis of facioscapulohumeral dystrophy. The underlying molecular mechanism by which Smchd1 functions in these instances remains unknown. Our genome-wide transcriptional and epigenetic analyses show that Smchd1 binds cis-regulatory elements, many of which coincide with CCCTC-binding factor (Ctcf) binding sites, for example, the clustered protocadherin (Pcdh) genes, where we show Smchd1 and Ctcf act in opposing ways. We provide biochemical and biophysical evidence that Smchd1-chromatin interactions are established through the homodimeric hinge domain of Smchd1 and, intriguingly, that the hinge domain also has the capacity to bind DNA and RNA. Our results suggest Smchd1 imparts epigenetic regulation via physical association with chromatin, which may antagonize Ctcf-facilitated chromatin interactions, resulting in coordinated transcriptional control.


Assuntos
Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Epigênese Genética , Genoma , Animais , Sítios de Ligação/genética , Western Blotting , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/metabolismo , Fator de Ligação a CCCTC , Células Cultivadas , Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica , Histonas/metabolismo , Masculino , Metilação , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/metabolismo , Ligação Proteica , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcriptoma/genética
4.
Epigenetics Chromatin ; 6(1): 19, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23819640

RESUMO

BACKGROUND: Smchd1 is an epigenetic modifier essential for X chromosome inactivation: female embryos lacking Smchd1 fail during midgestational development. Male mice are less affected by Smchd1-loss, with some (but not all) surviving to become fertile adults on the FVB/n genetic background. On other genetic backgrounds, all males lacking Smchd1 die perinatally. This suggests that, in addition to being critical for X inactivation, Smchd1 functions to control the expression of essential autosomal genes. RESULTS: Using genome-wide microarray expression profiling and RNA-seq, we have identified additional genes that fail X inactivation in female Smchd1 mutants and have identified autosomal genes in male mice where the normal expression pattern depends upon Smchd1. A subset of genes in the Snrpn imprinted gene cluster show an epigenetic signature and biallelic expression consistent with loss of imprinting in the absence of Smchd1. In addition, single nucleotide polymorphism analysis of expressed genes in the placenta shows that the Igf2r imprinted gene cluster is also disrupted, with Slc22a3 showing biallelic expression in the absence of Smchd1. In both cases, the disruption was not due to loss of the differential methylation that marks the imprint control region, but affected genes remote from this primary imprint controlling element. The clustered protocadherins (Pcdhα, Pcdhß, and Pcdhγ) also show altered expression levels, suggesting that their unique pattern of random combinatorial monoallelic expression might also be disrupted. CONCLUSIONS: Smchd1 has a role in the expression of several autosomal gene clusters that are subject to monoallelic expression, rather than being restricted to functioning uniquely in X inactivation. Our findings, combined with the recent report implicating heterozygous mutations of SMCHD1 as a causal factor in the digenically inherited muscular weakness syndrome facioscapulohumeral muscular dystrophy-2, highlight the potential importance of Smchd1 in the etiology of diverse human diseases.

5.
Mamm Genome ; 24(5-6): 206-17, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23636699

RESUMO

Observations of inherited phenotypes that cannot be explained solely through genetic inheritance are increasing. Evidence points to transmission of non-DNA molecules in the gamete as mediators of the phenotypes. However, in most cases it is unclear what the molecules are, with DNA methylation, chromatin proteins, and small RNAs being the most prominent candidates. From a screen to generate novel mouse mutants of genes involved in epigenetic reprogramming, we produced a DNA methyltransferase 3b allele that is missing exon 13. Mice that are homozygous for the mutant allele have smaller stature and reduced viability, with particularly high levels of female post-natal death. Reduced DNA methylation was also detected at telocentric repeats and the X-linked Hprt gene. However, none of the abnormal phenotypes or DNA methylation changes worsened with multiple generations of homozygous mutant inbreeding. This suggests that in our model the abnormalities are reset each generation and the processes of transgenerational epigenetic reprogramming are effective in preventing their inheritance.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Camundongos/genética , Alelos , Animais , Sequência de Bases , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Epigênese Genética , Éxons , Feminino , Homozigoto , Masculino , Camundongos/crescimento & desenvolvimento , Camundongos/metabolismo , Camundongos Transgênicos , Dados de Sequência Molecular , Linhagem , DNA Metiltransferase 3B
6.
BMC Cancer ; 12: 252, 2012 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-22708734

RESUMO

BACKGROUND: While it is now more than a decade since the first description of the gene mutation underlying the tumour predisposition syndrome multiple endocrine neoplasia type 1 (MEN1), the mechanism by which its protein product menin acts to prevent development of tumours is still poorly understood. METHODS: We undertook a genetic experiment to assess whether menin synergises with p53. Mice carrying various combinations of Men1 and Trp53 mutations were generated then survival and pathology assessed. RESULTS: While homozygous loss of Trp53 in mice resulted in early onset, aggressive tumours and profoundly reduced lifespan, heterozygous loss of either Trp53 or Men1 caused later onset disease, with a spectrum of tumours characteristic of each tumour suppressor gene. Loss of one copy of Men1 in animals also lacking both alleles of Trp53 did not exacerbate phenotype, based on survival, animal weight or sites of pathology, compared to Trp53 deletion alone. Dual heterozygous deletion of Men1 and Trp53 resulted in a small reduction in lifespan compared to the individual mutations, without new tumour sites. In the adrenal, we observed development of cortical tumours in dual heterozygous animals, as we have previously seen in Men1+/- animals, and there was loss of heterozygosity at the Men1 allele in these tumours. Median number of pathology observations per animal was increased in dual heterozygous animals compared with heterozygous loss of Trp53 alone. CONCLUSIONS: Simultaneous heterozygous deletion of Men1 in animals with either heterozygous or homozygous deletion of Trp53 did not result in formation of tumours at any new sites, implying additive rather than synergistic effects of these pathways. Mice that were Men1+/- in addition to Trp53+/- had tumours in endocrine as well as other sites, implying that increase in total tumour burden, at sites typically associated with either Men1 or Trp53 loss, contributed to the slight decrease in survival in Men1+/-: Trp53+/- animals in comparison with their littermates.


Assuntos
Transformação Celular Neoplásica/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Proteína Supressora de Tumor p53/genética , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/patologia , Animais , Peso Corporal , Transformação Celular Neoplásica/metabolismo , Genótipo , Camundongos , Camundongos Knockout , Neoplasia Endócrina Múltipla Tipo 1/metabolismo , Mutação , Neoplasias/genética , Neoplasias/mortalidade , Neoplasias/patologia , Pâncreas/metabolismo , Pâncreas/patologia , Proteína Supressora de Tumor p53/metabolismo
8.
Nat Struct Mol Biol ; 17(8): 948-54, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20657585

RESUMO

One X chromosome, selected at random, is silenced in each female mammalian cell. Xist encodes a noncoding RNA that influences the probability that the cis-linked X chromosome will be silenced. We found that the A-repeat, a highly conserved element within Xist, is required for the accumulation of spliced Xist RNA. In addition, the A-repeat is necessary for X-inactivation to occur randomly. In combination, our data suggest that normal Xist RNA processing is important in the regulation of random X-inactivation. We propose that modulation of Xist RNA processing may be part of the stochastic process that determines which X chromosome will be inactivated.


Assuntos
Proteínas Nucleares/metabolismo , Processamento Pós-Transcricional do RNA , RNA não Traduzido/genética , Proteínas de Ligação a RNA/metabolismo , Sequências Repetitivas de Ácido Nucleico/genética , Inativação do Cromossomo X/genética , Alelos , Animais , Sequência de Bases , Cromossomos de Mamíferos/metabolismo , Feminino , Células HeLa , Histonas/metabolismo , Humanos , Masculino , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Ligação Proteica , Processamento de Proteína Pós-Traducional , RNA Longo não Codificante , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA não Traduzido/química , Deleção de Sequência/genética , Fatores de Processamento de Serina-Arginina
9.
Pancreas ; 39(8): 1140-6, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20531244

RESUMO

OBJECTIVES: To identify gene expression alterations associated with insulinoma formation and progression in 2 mouse models of multiple endocrine neoplasia type 1. METHODS: Mice were killed at 12 or 16 months, and pancreatic islets were isolated by enzymatic and physical disruption. Islets were separated by size representing control, normal, hyperplastic, and adenomous islets. RNA was isolated from these islets and profiled on Sentrix Mouse-6 Expression version 1 BeadChips. Array data were analyzed in GeneSpring. RESULTS: One hundred and one genes that were significantly (P ≤ 0.05) altered in hyperplastic islets and insulinomas compared with normal islets were identified. Of these, 64 gene elements showed reduced messenger RNA levels and 37 gene elements had increased gene expression compared with control islets. Altered expression of 3 genes, namely, Gata6, Tspan8, and s100a8, was confirmed by quantitative reverse transcription-polymerase chain reaction, and aberrant levels of Tspan8 and Lmo2 protein measured by Western blot correlated with the changes in messenger RNA levels. CONCLUSIONS: These results suggest that alterations in gene expression of Gata6, Tspan8, S100a8, and Lmo2 may act via novel pathways that play functionally important roles in Men1-associated tumor progression.


Assuntos
Perfilação da Expressão Gênica , Insulinoma/genética , Neoplasia Endócrina Múltipla Tipo 1/genética , Neoplasias Pancreáticas/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Western Blotting , Calgranulina A/genética , Calgranulina A/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Progressão da Doença , Feminino , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Humanos , Insulinoma/etiologia , Insulinoma/metabolismo , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Proteínas com Domínio LIM , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Metaloproteínas/genética , Metaloproteínas/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasia Endócrina Múltipla Tipo 1/complicações , Neoplasia Endócrina Múltipla Tipo 1/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tetraspaninas
10.
Pigment Cell Melanoma Res ; 23(4): 564-70, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20518858

RESUMO

There has been uncertainty regarding the precise role that the pocket protein Rb1 plays in murine melanocyte homeostasis. It has been reported that the TAT-Cre mediated loss of exon 19 from a floxed Rb1 allele causes melanocyte apoptosis in vivo and in vitro. This is at variance with other findings showing, either directly or indirectly, that Rb1 loss in melanocytes has no noticeable effect in vivo, but in vitro leads to a semi-transformed phenotype. In this study, we show that Rb1-null melanocytes lacking exon 19 do not undergo apoptosis and survive both in vitro and in vivo, irrespective of the developmental stage at which Cre-mediated ablation of the exon occurs. Further, Rb1 loss has no serious long-term ramifications on melanocyte homeostasis in vivo, with Rb1-null melanocytes being detected in the skin after numerous hair cycles, inferring that the melanocyte stem cell population carrying the Cre-mediated deletion is maintained. Consequently, whilst Rb1 loss in the melanocyte is able to alter cellular behaviour in vitro, it appears inconsequential with respect to melanocyte homeostasis in the mouse skin.


Assuntos
Cabelo/metabolismo , Homeostase , Melanócitos/metabolismo , Proteína do Retinoblastoma/deficiência , Pele/metabolismo , Animais , Cabelo/patologia , Camundongos , Camundongos Knockout , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Pele/patologia
11.
Neoplasia ; 12(3): 235-43, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20234817

RESUMO

Using a Cre/loxP system, we have determined the phenotypic consequences attributable to in vivo deletion of both Rb1 and Trp53 in the mouse adrenal medulla. The coablation of these two tumor suppressor genes during embryogenesis did not disrupt adrenal gland development but resulted in the neoplastic transformation of the neural crest-derived adrenal medulla, yielding pheochromocytomas (PCCs) that developed with complete penetrance and were inevitably bilateral. Despite their typically benign status, these PCCs had profound ramifications on mouse vitality, with effected mice having a median survival of only 121 days. Evaluation of these PCCs by both immunohistochemistry and electron microscopy revealed that most Rb1(-/-):Trp53(-/-) chromaffin cells possessed atypical chromagenic vesicles that did not seem capable of appropriately storing synthesized catecholamines. The structural remodeling of the heart in mice harboring Rb1(-/-):Trp53(-/-) PCCs suggests that the mortality of these mice may be attributable to the inappropriate release of catecholamines from the mutated adrenal chromaffin cells. On the basis of the collective data from Rb1 and Trp53 knockout mouse models, it seems that the conversion of Rb1 loss-driven adrenal medulla hyperplasia to PCC can be greatly enhanced by the compound loss of Trp53, whereas the loss of Trp53 alone is generally ineffectual on adrenal chromaffin cell homeostasis. Consequently, the Trp53 tumor suppressor gene is an efficient genetic modifier of Rb1 loss in the development of PCC, and their compound loss in the adrenal medulla has a profound impact on both cellular homeostasis and animal vitality.


Assuntos
Neoplasias das Glândulas Suprarrenais/patologia , Genes p53/fisiologia , Feocromocitoma/patologia , Proteína do Retinoblastoma/fisiologia , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/metabolismo , Animais , Catecolaminas/metabolismo , Feminino , Técnicas Imunoenzimáticas , Integrases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Feocromocitoma/genética , Feocromocitoma/metabolismo , Transgenes/fisiologia
12.
Biol Reprod ; 81(5): 921-32, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19571264

RESUMO

An estimated 25%-40% of infertile men have idiopathic infertility associated with deficient sperm numbers and quality. Here, we identify the membrane-anchored serine protease PRSS21, also known as testisin, to be a novel proteolytic factor that directs epididymal sperm cell maturation and sperm-fertilizing ability. PRSS21-deficient spermatozoa show decreased motility, angulated and curled tails, fragile necks, and dramatically increased susceptibility to decapitation. These defects reflect aberrant maturation during passage through the epididymis, because histological and electron microscopic structural analyses showed an increased tendency for curled and detached tails as spermatozoa transit from the corpus to the cauda epididymis. Cauda epididymal spermatozoa deficient in PRSS21 fail to mount a swelling response when exposed to hypotonic conditions, suggesting an impaired ability to respond to osmotic challenges facing maturing spermatozoa in the female reproductive tract. These data suggest that aberrant regulation of PRSS21 may underlie certain secondary male infertility syndromes, such as "easily decapitated" spermatozoa in humans.


Assuntos
Fertilização/fisiologia , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Maturação do Esperma/fisiologia , Motilidade dos Espermatozoides/fisiologia , Espermatozoides/citologia , Animais , Western Blotting , Contagem de Células , Forma Celular , Sobrevivência Celular , Copulação/fisiologia , Feminino , Fertilização in vitro , Proteínas Ligadas por GPI , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Fosforilação , Serina Endopeptidases/genética , Espermatozoides/metabolismo , Coloração e Rotulagem
14.
Int J Cancer ; 124(5): 1122-32, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19058182

RESUMO

Heterozygous disruption of the Men1 gene predisposes mice to the development of multiple endocrine tumors, accurately mimicking the human MEN1 cancer predisposition syndrome. Additionally, Men1(+/-) mice frequently develop sex cord adenomas. The mechanism underlying the susceptibility of these mice to sex cord tumor development has not been fully determined, but data suggest it may involve transcriptional regulation of key growth promoting/repressing genes. To identify potential menin-regulated genes that may be important for tumor suppression in sex cord cells, we compared the global gene expression profiles of testis and ovary adenomas with other endocrine tumors of the pancreas and pituitary from Men1 heterozygous mice and with control tissues. Gonadal tumors clustered separately from pancreas and pituitary tumors with only a few genes (e.g., Cdkn2c) commonly dysregulated in all tumor types. Testis and ovary tumors displayed a higher level of transcriptional similarity to each other than they did to their respective control tissues. Among genes that had decreased expression in tumors was significant over-representation of genes associated with the TGF-beta, hedgehog and Wnt signaling, indicating that loss of menin function affects these pathways at the level of transcription. Aberrant protein expression in Leydig and granulosa cells of 2 transcriptionally dysregulated gene products, Gata6 and Csf1r were confirmed by immunohistochemistry. We propose that sex cord tumor susceptibility in Men1(+/-) mice involves deregulated cell proliferation due to dysregulation of multiple cell growth regulating genes including: reduced Cdkn2c transcription, loss of TGF-beta pathway tumor suppressor function (e.g., Gata6) and transcriptional activation of Csf1r.


Assuntos
Fator de Transcrição GATA6/análise , Perfilação da Expressão Gênica , Proteínas Proto-Oncogênicas/fisiologia , Receptor de Fator Estimulador de Colônias de Macrófagos/análise , Tumores do Estroma Gonadal e dos Cordões Sexuais/etiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Análise por Conglomerados , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas/genética
15.
J Natl Cancer Inst ; 100(21): 1519-29, 2008 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-18957670

RESUMO

BACKGROUND: X chromosome inactivation, which silences gene expression from one of the two X chromosomes in females, is usually random. Skewed X inactivation has been implicated in both the expression and the suppression of X-linked disease phenotypes and has been reported to occur more frequently in breast and ovarian cancer patients, including BRCA1 or BRCA2 mutation carriers, than in control subjects. METHODS: We assessed the pattern of X chromosome inactivation using methylation-specific polymerase chain reaction amplification of the exon 1 microsatellite region of the X-linked androgen receptor (AR) gene in DNA from blood samples obtained from control subjects without a personal history of breast or ovarian cancer (n = 735), ovarian cancer patients (n = 313), familial breast cancer patients who did not carry mutations in BRCA1 or BRCA2 (n = 235), and affected and unaffected carriers of mutations in BRCA1 (n = 260) or BRCA2 (n = 63). We defined the pattern of X chromosome inactivation as skewed when the same X chromosome was active in at least 90% of cells. The association between skewed X inactivation and disease and/or BRCA mutation status was assessed by logistic regression analysis. The association between skewed X inactivation and age at cancer diagnosis was assessed by Cox proportional hazards regression analysis. All statistical tests were two-sided. RESULTS: The age-adjusted frequency of skewed X inactivation was not statistically significantly higher in ovarian cancer or familial breast cancer case subjects compared with control subjects. Skewed X inactivation was higher in BRCA1 mutation carriers than in control subjects (odds ratio [OR] = 2.7, 95% confidence interval [CI] = 1.1 to 6.2; P = .02), particularly among unaffected women (OR = 6.1, 95% CI = 1.5 to 31.8; P = .005). Among BRCA1 mutation carriers, those with skewed X inactivation were older at diagnosis of breast or ovarian cancer than those without skewed X inactivation (hazard ratio [HR] of breast or ovarian cancer = 0.37, 95% CI = 0.14 to 0.95; P = .04). Among BRCA2 mutation carriers, skewed X inactivation also occurred more frequently in unaffected carriers than in those diagnosed with breast or ovarian cancer (OR = 5.2, 95% CI = 0.5 to 28.9; P = .08) and was associated with delayed age at onset (HR = 0.59, 95% CI = 0.37 to 0.94; P = .03). CONCLUSIONS: Skewed X inactivation occurs at an increased frequency in BRCA1 (and possibly BRCA2) mutation carriers compared with control subjects and is associated with a statistically significant increase in age at diagnosis of breast and ovarian cancer.


Assuntos
Neoplasias da Mama/genética , Cromossomos Humanos X , Genes BRCA1 , Mutação , Neoplasias Ovarianas/genética , Receptores Androgênicos/genética , Inativação do Cromossomo X , Adolescente , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Estudos de Casos e Controles , Metilação de DNA , Primers do DNA , DNA de Neoplasias/genética , Feminino , Genes BRCA2 , Genes Ligados ao Cromossomo X , Predisposição Genética para Doença , Genótipo , Heterozigoto , Humanos , Estimativa de Kaplan-Meier , Modelos Logísticos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Razão de Chances , Reação em Cadeia da Polimerase/métodos , Modelos de Riscos Proporcionais , Regulação para Cima
16.
Neoplasia ; 10(11): 1213-21, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18953430

RESUMO

Signaling events leading to Schwann cell tumor initiation have been extensively characterized in the context of neurofibromatosis (NF). Similar tumors are also observed in patients with the endocrine neoplasia syndrome Carney complex, which results from inactivating mutations in PRKAR1A. Loss of PRKAR1A causes enhanced protein kinase A activity, although the pathways leading to tumorigenesis are not well characterized. Tissue-specific ablation of Prkar1a in neural crest precursor cells (TEC3KO mice) causes schwannomas with nearly 80% penetrance by 10 months. These heterogeneous neoplasms were clinically characterized as genetically engineered mouse schwannomas, grades II and III. At the molecular level, analysis of the tumors revealed almost complete loss of both NF proteins, despite the fact that transcript levels were increased, implying posttranscriptional regulation. Although Erk and Akt signaling are typically enhanced in NF-associated tumors, we observed no activation of either of these pathways in TEC3KO tumors. Furthermore, the small G proteins Ras, Rac1, and RhoA are all known to be involved with NF signaling. In TEC3KO tumors, all three molecules showed modest increases in total protein, but only Rac1 showed significant activation. These data suggest that dysregulated protein kinase A activation causes tumorigenesis through pathways that overlap but are distinct from those described in NF tumorigenesis.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Neurilemoma/metabolismo , Neurofibromina 1/metabolismo , Neurofibromina 2/metabolismo , Animais , Western Blotting , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Genes da Neurofibromatose 1 , Genes da Neurofibromatose 2 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neurilemoma/genética , Reação em Cadeia da Polimerase , Células de Schwann/metabolismo , Transdução de Sinais
17.
Genesis ; 46(10): 553-61, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18802966

RESUMO

Eph receptor tyrosine kinases (RTKs) are a highly conserved family of signaling proteins with functions in cellular migration, adhesion, apoptosis, and proliferation during both adult and embryonic life. Here, we describe a knock-in mouse in which EphA1 expression is disrupted via the insertion of an internal ribosome entry site (IRES)-human placental alkaline phosphatase (ALPP) reporter cassette into exon II of the EphA1 gene. This was shown to successfully knockout expression of endogenous EphA1 and enforce expression of the ALPP reporter by the EphA1 promoter. Staining for the ALPP reporter protein demonstrated an epithelially restricted expression pattern in mouse tissues. In EphA1 null mice, two separate phenotypes were identified: abnormal tail development manifesting as a kinky tail was found in approximately 80% of homozygous adults. A second, distinct abnormality present in approximately 18% of females was characterized by imperforate uterovaginal development with hydrometrocolpos and caused by a resistance of cells to apoptosis during reproductive tract canalization. These results indicate a possible role for EphA1 in tissue patterning and hormone-induced apoptotic processes.


Assuntos
Genes Reporter , Receptor EphA1/genética , Fosfatase Alcalina , Animais , Apoptose/genética , Padronização Corporal/genética , Efrina-A1/metabolismo , Feminino , Proteínas Ligadas por GPI , Técnicas de Introdução de Genes , Humanos , Isoenzimas/genética , Masculino , Camundongos , Camundongos Knockout , Receptor EphA1/fisiologia , Cauda/anormalidades , Cauda/citologia , Cauda/enzimologia , Útero/anormalidades , Útero/citologia , Útero/enzimologia , Vagina/anormalidades , Vagina/citologia , Vagina/enzimologia
18.
Nat Genet ; 40(5): 663-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18425126

RESUMO

X-chromosome inactivation is the mammalian dosage compensation mechanism by which transcription of X-linked genes is equalized between females and males. In an N-ethyl-N-nitrosourea (ENU) mutagenesis screen on mice for modifiers of epigenetic reprogramming, we identified the MommeD1 (modifier of murine metastable epialleles) mutation as a semidominant suppressor of variegation. MommeD1 shows homozygous female-specific mid-gestation lethality and hypomethylation of the X-linked gene Hprt1, suggestive of a defect in X inactivation. Here we report that the causative point mutation lies in a previously uncharacterized gene, Smchd1 (structural maintenance of chromosomes hinge domain containing 1). We find that SmcHD1 is not required for correct Xist expression, but localizes to the inactive X and has a role in the maintenance of X inactivation and the hypermethylation of CpG islands associated with the inactive X. This finding links a group of proteins normally associated with structural aspects of chromosome biology with epigenetic gene silencing.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Inativação Gênica , Inativação do Cromossomo X , Cromossomo X/metabolismo , Animais , Proteínas Cromossômicas não Histona/análise , Proteínas Cromossômicas não Histona/genética , Ilhas de CpG , Metilação de DNA , Fibroblastos/ultraestrutura , Camundongos , Mutação Puntual , RNA Longo não Codificante , RNA não Traduzido/metabolismo , Cromossomo X/química , Cromossomo X/genética
19.
Int J Cancer ; 121(4): 776-83, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17455252

RESUMO

Although the identification of menin-interacting partners and other evidence support a role for menin, the multiple endocrine neoplasia type 1 gene (MEN1) product, in regulating gene expression, little is known about the cellular pathways dysregulated by menin loss during tumorigenesis. The mouse models of MEN1 accurately mimic the human syndrome and provide an opportunity to assess the transcriptional effects of Men1 deletion in different endocrine tumor types to identify common pathway aberrations underlying tumorigenesis in MEN1-affected tissues. We compared the global gene expression profiles of pituitary adenomas and pancreatic islet tumors with control tissues from wild-type littermates. Amongst the 551 differentially expressed genes was significant over-representation of genes associated with chromatin remodelling, transcription and cell cycling, including some genes known to encode menin-binding partners, e.g., Rhox5 and Mll1. Consistent with increased cell-cycle transition from G1 to S phase was an elevation of Cdc7 expression in the tumors, which was confirmed by qRT-PCR using independent samples. In support of previous findings in islet tumors, we found down-regulation of the cell-cycle regulator, p18, in both the pancreatic islet and pituitary adenomas, suggesting that reduced p18 levels may be important for Men1-related tumorigenesis in multiple tissues. Surprisingly, we identified increased p16 transcript in pancreatic islet and pituitary tumors. This was accompanied by increased cytoplasmic localization p16 protein in tumor cells. The specific genes and general pathways we have found to be commonly dysregulated in MEN1 tumors, provide a platform for determining their roles in endocrine tumorigenesis.


Assuntos
Ciclo Celular , Montagem e Desmontagem da Cromatina , Perfilação da Expressão Gênica , Neoplasias Experimentais/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Transcrição Gênica , Animais , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Knockout , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Pancreáticas/genética , Neoplasias Hipofisárias/genética , Reprodutibilidade dos Testes
20.
Int J Cancer ; 120(2): 259-67, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17044021

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) is an inherited cancer predisposition syndrome typified by development of tumors in parathyroid, pituitary and endocrine pancreas, as well as less common sites including both endocrine and nonendocrine organs. Deletion or mutation of the tumor suppressor gene MEN1 on chromosome 11 has been identified in many cases of MEN1 as well as in sporadic tumors. The molecular biology of menin, the protein encoded by MEN1, remains poorly understood. Here we describe a mouse model of MEN1 in which tumors were seen in pancreatic islets, pituitary, thyroid and parathyroid, adrenal glands, testes and ovaries. The observed tumor spectrum therefore includes types commonly seen in MEN1 patients and additional types. Pancreatic pathology was most common, evident in over 80% of animals, while other tumor types developed with lower frequency and generally later onset. Tumors of multiple endocrine organs were observed frequently, but progression to carcinoma and metastasis were not evident. Tumors in all sites showed loss of heterozygosity at the Men1 locus, though the frequency in testicular tumors was only 36%, indicating that a different molecular mechanism of tumorigenesis occurs in those Leydig tumors that do not show loss of the normal Men1 allele. Menin expression was below the level of detection in ovary, thyroid and testis, but loss of nuclear menin immunoreactivity was observed uniformly in all pancreatic islet adenomas and in some hyperplastic islet cells, suggesting that complete loss of Men1 is a critical point in islet tumor progression in this model.


Assuntos
Adenoma/patologia , Modelos Animais de Doenças , Neoplasias das Glândulas Endócrinas/patologia , Camundongos/genética , Neoplasia Endócrina Múltipla Tipo 1/patologia , Proteínas Proto-Oncogênicas/genética , Adenoma/química , Adenoma/genética , Animais , DNA de Neoplasias/análise , Neoplasias das Glândulas Endócrinas/química , Neoplasias das Glândulas Endócrinas/genética , Éxons/genética , Feminino , Genes Supressores de Tumor , Masculino , Neoplasia Endócrina Múltipla Tipo 1/química , Neoplasia Endócrina Múltipla Tipo 1/genética , Iniciação Traducional da Cadeia Peptídica/genética , Proteínas Proto-Oncogênicas/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...