Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Prostate ; 80(6): 491-499, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32068909

RESUMO

BACKGROUND: Prostate cancer (PC) research has relied heavily on patient-derived cell lines, which may be used for in vitro (two-dimensional [2D]) studies or cultivated as three-dimensional (3D) xenografts in mice. These approaches are likely to have differential impacts on cell phenotypes, with implications for experimental outcomes. Therefore, defining and comparing the transcriptional signatures associated with 2D and 3D approaches may be useful for designing experiments and interpreting research results. METHODS: In this study, LNCaP, VCaP, and 22Rv1 human PC cells were either cultivated in monolayers or as xenografts in NOD SCID mice, and their gene transcription profiles were quantitated and compared using microarray and real-time polymerase chain reaction techniques. Immunohistochemistry was used to evaluate protein expression in cancer cell xenografts. RESULTS: Comparisons of gene expression profiles of tumor cells grown in 2D vs 3D environments identified gene sets featuring similar expression patterns in all three cancer cell lines and unique transcriptional signatures associated with 3D vs 2D growth. Pathways related to cell-cell interactions, differentiation, and the extracellular matrix were enriched in 3D conditions. Immunohistochemical analyses confirmed that gene upregulation in xenografts occurred in implanted cancer cells and not in mouse stromal cells. Cultivating cells in vitro in the presence of mouse, rather than bovine serum failed to elicit the gene transcription profile observed in xenografts, further supporting the hypothesis that this profile reflects 3D growth and enhanced microenvironmental interactions, rather than exposure to species-specific serum factors. CONCLUSIONS: Overall, these findings define the expression profiles observed in PC cells cultivated in 2D monolayers and in 3D xenografts, highlighting differentially regulated pathways in each setting and providing information for interpreting research results in model systems.


Assuntos
Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Xenoenxertos , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Transcriptoma , Células Tumorais Cultivadas , Microambiente Tumoral/genética
2.
Cereb Cortex ; 28(2): 493-509, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28031177

RESUMO

Mice that are constitutively null for the zinc finger doublesex and mab-3 related (Dmrt) gene, Dmrt5/Dmrta2, show a variety of patterning abnormalities in the cerebral cortex, including the loss of the cortical hem, a powerful cortical signaling center. In conditional Dmrt5 gain of function and loss of function mouse models, we generated bidirectional changes in the neocortical area map without affecting the hem. Analysis indicated that DMRT5, independent of the hem, directs the rostral-to-caudal pattern of the neocortical area map. Thus, DMRT5 joins a small number of transcription factors shown to control directly area size and position in the neocortex. Dmrt5 deletion after hem formation also reduced hippocampal size and shifted the position of the neocortical/paleocortical boundary. Dmrt3, like Dmrt5, is expressed in a gradient across the cortical primordium. Mice lacking Dmrt3 show cortical patterning defects akin to but milder than those in Dmrt5 mutants, perhaps in part because Dmrt5 expression increases in the absence of Dmrt3. DMRT5 upregulates Dmrt3 expression and negatively regulates its own expression, which may stabilize the level of DMRT5. Together, our findings indicate that finely tuned levels of DMRT5, together with DMRT3, regulate patterning of the cerebral cortex.


Assuntos
Desenvolvimento Embrionário/fisiologia , Hipocampo/metabolismo , Neocórtex/metabolismo , Fatores de Transcrição/biossíntese , Animais , Hipocampo/embriologia , Hipocampo/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neocórtex/embriologia , Neocórtex/crescimento & desenvolvimento , Neurogênese/fisiologia
3.
Nat Rev Genet ; 13(3): 163-74, 2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22310892

RESUMO

Most animals reproduce sexually, but the genetic and molecular mechanisms that determine the eventual sex of each embryo vary remarkably. DM domain genes, which are related to the insect gene doublesex, are integral to sexual development and its evolution in many metazoans. Recent studies of DM domain genes reveal mechanisms by which new sexual dimorphisms have evolved in invertebrates and show that one gene, Dmrt1, was central to multiple evolutionary transitions between sex-determining mechanisms in vertebrates. In addition, Dmrt1 coordinates a surprising array of distinct cell fate decisions in the mammalian gonad and even guards against transdifferentiation of male cells into female cells in the adult testis.


Assuntos
Evolução Biológica , Genitália/embriologia , Células Germinativas/fisiologia , Processos de Determinação Sexual , Fatores de Transcrição/fisiologia , Adulto , Animais , Feminino , Humanos , Masculino
4.
Nature ; 476(7358): 101-4, 2011 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-21775990

RESUMO

Sex in mammals is determined in the fetal gonad by the presence or absence of the Y chromosome gene Sry, which controls whether bipotential precursor cells differentiate into testicular Sertoli cells or ovarian granulosa cells. This pivotal decision in a single gonadal cell type ultimately controls sexual differentiation throughout the body. Sex determination can be viewed as a battle for primacy in the fetal gonad between a male regulatory gene network in which Sry activates Sox9 and a female network involving WNT/ß-catenin signalling. In females the primary sex-determining decision is not final: loss of the FOXL2 transcription factor in adult granulosa cells can reprogram granulosa cells into Sertoli cells. Here we show that sexual fate is also surprisingly labile in the testis: loss of the DMRT1 transcription factor in mouse Sertoli cells, even in adults, activates Foxl2 and reprograms Sertoli cells into granulosa cells. In this environment, theca cells form, oestrogen is produced and germ cells appear feminized. Thus Dmrt1 is essential to maintain mammalian testis determination, and competing regulatory networks maintain gonadal sex long after the fetal choice between male and female. Dmrt1 and Foxl2 are conserved throughout vertebrates and Dmrt1-related sexual regulators are conserved throughout metazoans. Antagonism between Dmrt1 and Foxl2 for control of gonadal sex may therefore extend beyond mammals. Reprogramming due to loss of Dmrt1 also may help explain the aetiology of human syndromes linked to DMRT1, including disorders of sexual differentiation and testicular cancer.


Assuntos
Caracteres Sexuais , Processos de Determinação Sexual/fisiologia , Diferenciação Sexual/fisiologia , Testículo/metabolismo , Fatores de Transcrição/metabolismo , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Transdiferenciação Celular , Feminino , Feminização/genética , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Masculino , Camundongos , Modelos Biológicos , Ovário/citologia , Ovário/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOX9/metabolismo , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Processos de Determinação Sexual/genética , Diferenciação Sexual/genética , Testículo/citologia , Células Tecais/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
5.
Cereb Cortex ; 23(11): 2552-67, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22923088

RESUMO

Regional patterning of the cerebral cortex is initiated by morphogens secreted by patterning centers that establish graded expression of transcription factors within cortical progenitors. Here, we show that Dmrt5 is expressed in cortical progenitors in a high-caudomedial to low-rostrolateral gradient. In its absence, the cortex is strongly reduced and exhibits severe abnormalities, including agenesis of the hippocampus and choroid plexus and defects in commissural and thalamocortical tracts. Loss of Dmrt5 results in decreased Wnt and Bmp in one of the major telencephalic patterning centers, the dorsomedial telencephalon, and in a reduction of Cajal-Retzius cells. Expression of the dorsal midline signaling center-dependent transcription factors is downregulated, including Emx2, which promotes caudomedial fates, while the rostral determinant Pax6, which is inhibited by midline signals, is upregulated. Consistently, Dmrt5(-/-) brains exhibit patterning defects with a dramatic reduction of the caudomedial cortex. Dmrt5 is increased upon the activation of Wnt signaling and downregulated in Gli3(xt/xt) mutants. We conclude that Dmrt5 is a novel Wnt-dependent transcription factor required for early cortical development and that it may regulate initial cortical patterning by promoting dorsal midline signaling center formation and thereby helping to establish the graded expression of the other transcription regulators of cortical identity.


Assuntos
Córtex Cerebral/embriologia , Fatores de Transcrição/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Córtex Cerebral/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição/genética , Proteínas Wnt/metabolismo
6.
ACS Med Chem Lett ; 11(6): 1236-1243, 2020 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-32551006

RESUMO

A series of PI3Kß selective inhibitors derived from a novel 4-(1H-benzo[d]imidazol-1-yl)quinoline chemotype has been rationally designed. Crucial to achieving the desired selectivity over the other class I PI3K isoforms, including the challenging δ-isoform, was the identification of a subset of substituted pyridine hinge binders. This work led to the discovery of (P)-14, a highly selective and orally bioavailable PI3Kß inhibitor displaying an excellent pharmacokinetic profile in addition to great cellular potency in various PTEN-deficient tumor cell lines. Results from a dog toxicology study revealing structure-related, off-target ocular toxicity are also briefly discussed.

7.
Curr Biol ; 14(16): 1416-24, 2004 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-15324658

RESUMO

BACKGROUND: Many different environmental and genetic sex-determination mechanisms are found in nature. Closely related species can use different master sex-determination switches, suggesting that these developmental pathways can evolve very rapidly. Previous cytological studies suggest that recently diverged species of stickleback fish have different sex chromosome complements. Here, we investigate the genetic and chromosomal mechanisms that underlie sex determination in the threespine stickleback (Gasterosteus aculeatus). RESULTS: Genome-wide linkage mapping identifies a single chromosome region at the distal end of linkage group (LG) 19, which controls male or female sexual development in threespine sticklebacks. Although sex chromosomes are not cytogenetically visible in this species, several lines of evidence suggest that LG 19 is an evolving sex chromosome system, similar to the XX female/XY male system in many other species: (1) males are consistently heterozygous for unique alleles in this region; (2) recombination between loci linked to the sex-determination region is reduced in male meiosis relative to female meiosis; (3) sequence analysis of X- and Y-specific bacterial artificial chromosome (BAC) clones from the sex-determination region reveals many sequence differences between the X- and Y-specific clones; and (4) the Y chromosome has accumulated transposable elements and local duplications. CONCLUSIONS: Taken together, our data suggest that threespine sticklebacks have a simple chromosomal mechanism for sex determination based on a nascent Y chromosome that is less than 10 million years old. Further analysis of the stickleback system will provide an exciting window into the evolution of sex-determination pathways and sex chromosomes in vertebrates.


Assuntos
Evolução Molecular , Processos de Determinação Sexual , Smegmamorpha/genética , Cromossomo Y/genética , Animais , Sequência de Bases , Mapeamento Cromossômico , Cromossomos Artificiais Bacterianos , Análise por Conglomerados , Cruzamentos Genéticos , Primers do DNA , Bases de Dados de Ácidos Nucleicos , Feminino , Masculino , Dados de Sequência Molecular , Filogenia , Recombinação Genética , Sequências Repetitivas de Ácido Nucleico/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência
8.
Dev Cell ; 29(5): 511-520, 2014 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-24856513

RESUMO

Mammalian sex determination initiates in the fetal gonad with specification of bipotential precursor cells into male Sertoli cells or female granulosa cells. This choice was long presumed to be irreversible, but genetic analysis in the mouse recently revealed that sexual fates must be maintained throughout life. Somatic cells in the testis or ovary, even in adults, can be induced to transdifferentiate to their opposite-sex equivalents by loss of a single transcription factor, DMRT1 in the testis or FOXL2 in the ovary. Here, we investigate what mechanism DMRT1 prevents from triggering transdifferentiation. We find that DMRT1 blocks testicular retinoic acid (RA) signaling from activating genes normally involved in female sex determination and ovarian development and show that inappropriate activation of these genes can drive sexual transdifferentiation. By preventing activation of potential feminizing genes, DMRT1 allows Sertoli cells to participate in RA signaling, which is essential for reproduction, without being sexually reprogrammed.


Assuntos
Transdiferenciação Celular/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Ovário/citologia , Retinoides/farmacologia , Células de Sertoli/citologia , Testículo/citologia , Fatores de Transcrição/metabolismo , Animais , Western Blotting , Feminino , Imunofluorescência , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/genética , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ovário/efeitos dos fármacos , Ovário/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores do Ácido Retinoico/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOX9/metabolismo , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Processos de Determinação Sexual/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos
9.
Dev Cell ; 19(4): 612-24, 2010 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-20951351

RESUMO

The switch from mitosis to meiosis is a unique feature of germ cell development. In mammals, meiotic initiation requires retinoic acid (RA), which activates meiotic inducers, including Stra8, but how the switch to meiosis is controlled in male germ cells (spermatogonia) remains poorly understood. Here we examine the role of the Doublesex-related transcription factor DMRT1 in adult spermatogenesis using conditional gene targeting in the mouse. Loss of Dmrt1 causes spermatogonia to precociously exit the spermatogonial program and enter meiosis. Therefore, DMRT1 determines whether male germ cells undergo mitosis and spermatogonial differentiation or meiosis. Loss of Dmrt1 in spermatogonia also disrupts cyclical gene expression in Sertoli cells. DMRT1 acts in spermatogonia to restrict RA responsiveness, directly repress Stra8 transcription, and activate transcription of the spermatogonial differentiation factor Sohlh1, thereby preventing meiosis and promoting spermatogonial development. By coordinating spermatogonial development and mitotic amplification with meiosis, DMRT1 allows abundant, continuous production of sperm.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Drosophila/química , Meiose/genética , Mitose/genética , Homologia de Sequência de Aminoácidos , Espermatozoides/citologia , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas Adaptadoras de Transdução de Sinal , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Cinética , Masculino , Meiose/efeitos dos fármacos , Camundongos , Mitose/efeitos dos fármacos , Mutação/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Proteínas/genética , Células de Sertoli/citologia , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Espermatogênese/efeitos dos fármacos , Espermatogênese/genética , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Espermatogônias/metabolismo , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Tretinoína/farmacologia
10.
Eukaryot Cell ; 5(4): 712-22, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16607018

RESUMO

Endoplasmic reticulum-associated degradation (ERAD) mediates the turnover of short-lived and misfolded proteins in the ER membrane or lumen. In spite of its important role, only subtle growth phenotypes have been associated with defects in ERAD. We have discovered that the ERAD proteins Ubc7 (Qri8), Cue1, and Doa10 (Ssm4) are required for growth of yeast that express high levels of the sterol biosynthetic enzyme, 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR). Interestingly, the observed growth defect was exacerbated at low temperatures, producing an HMGR-dependent cold sensitivity. Yeast strains lacking UBC7, CUE1, or DOA10 also assembled aberrant karmellae (ordered arrays of membranes surrounding the nucleus that assemble when HMGR is expressed at high levels). However, rather than reflecting the accumulation of abnormal karmellae, the cold sensitivity of these ERAD mutants was due to increased HMGR catalytic activity. Mutations that compromise proteasomal function also resulted in cold-sensitive growth of yeast with elevated HMGR, suggesting that improper degradation of ERAD targets might be responsible for the observed cold-sensitive phenotype. However, the essential ERAD targets were not the yeast HMGR enzymes themselves. The sterol metabolite profile of ubc7Delta cells was altered relative to that of wild-type cells. Since sterol levels are known to regulate membrane fluidity, the viability of ERAD mutants expressing normal levels of HMGR was examined at low temperatures. Cells lacking UBC7, CUE1, or DOA10 were cold sensitive, suggesting that these ERAD proteins have a role in cold adaptation, perhaps through effects on sterol biosynthesis.


Assuntos
Aclimatação/fisiologia , Proteínas de Transporte/fisiologia , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/fisiologia , Fosfoproteínas Fosfatases/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/fisiologia , Esteróis/biossíntese , Enzimas de Conjugação de Ubiquitina/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Proteínas de Transporte/genética , Temperatura Baixa , Deleção de Genes , Proteínas de Membrana/genética , Fosfoproteínas Fosfatases/genética , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Esteróis/análise , Enzimas Ativadoras de Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Ubiquitina-Proteína Ligases/genética , Regulação para Cima
11.
Yeast ; 20(10): 881-92, 2003 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-12868057

RESUMO

Increased levels of HMG-CoA reductase induce cell type- and isozyme-specific proliferation of the endoplasmic reticulum. In yeast, the ER proliferations induced by Hmg1p consist of nuclear-associated stacks of smooth ER membranes known as karmellae. To identify genes required for karmellae assembly, we compared the composition of populations of homozygous diploid S. cerevisiae deletion mutants following 20 generations of growth with and without karmellae. Using an initial population of 1,557 deletion mutants, 120 potential mutants were identified as a result of three independent experiments. Each experiment produced a largely non-overlapping set of potential mutants, suggesting that differences in specific growth conditions could be used to maximize the comprehensiveness of similar parallel analysis screens. Only two genes, UBC7 and YAL011W, were identified in all three experiments. Subsequent analysis of individual mutant strains confirmed that each experiment was identifying valid mutations, based on the mutant's sensitivity to elevated HMG-CoA reductase and inability to assemble normal karmellae. The largest class of HMG-CoA reductase-sensitive mutations was a subset of genes that are involved in chromatin structure and transcriptional regulation, suggesting that karmellae assembly requires changes in transcription or that the presence of karmellae may interfere with normal transcriptional regulation.


Assuntos
Retículo Endoplasmático/genética , Genes Fúngicos/genética , Hidroximetilglutaril-CoA Redutases/genética , Saccharomyces cerevisiae/genética , DNA Fúngico/genética , DNA Fúngico/metabolismo , Retículo Endoplasmático/metabolismo , Deleção de Genes , Regulação Fúngica da Expressão Gênica , Genes Fúngicos/fisiologia , Hidroximetilglutaril-CoA Redutases/metabolismo , Mutagênese , Análise de Sequência com Séries de Oligonucleotídeos , Saccharomyces cerevisiae/metabolismo , Transformação Genética/genética , Transformação Genética/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA