Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Exp Clin Cancer Res ; 43(1): 165, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38877560

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is the most lethal cancer with an aggressive metastatic phenotype and very poor clinical prognosis. Interestingly, a lower occurrence of PDAC has been described in individuals with severe and long-standing asthma. Here we explored the potential link between PDAC and the glucocorticoid (GC) budesonide, a first-line therapy to treat asthma. METHODS: We tested the effect of budesonide and the classical GCs on the morphology, proliferation, migration and invasiveness of patient-derived PDAC cells and pancreatic cancer cell lines, using 2D and 3D cultures in vitro. Furthermore, a xenograft model was used to investigate the effect of budesonide on PDAC tumor growth in vivo. Finally, we combined genome-wide transcriptome analysis with genetic and pharmacological approaches to explore the mechanisms underlying budesonide activities in the different environmental conditions. RESULTS: We found that in 2D culture settings, high micromolar concentrations of budesonide reduced the mesenchymal invasive/migrating features of PDAC cells, without affecting proliferation or survival. This activity was specific and independent of the Glucocorticoid Receptor (GR). Conversely, in a more physiological 3D environment, low nanomolar concentrations of budesonide strongly reduced PDAC cell proliferation in a GR-dependent manner. Accordingly, we found that budesonide reduced PDAC tumor growth in vivo. Mechanistically, we demonstrated that the 3D environment drives the cells towards a general metabolic reprogramming involving protein, lipid, and energy metabolism (e.g., increased glycolysis dependency). This metabolic change sensitizes PDAC cells to the anti-proliferative effect of budesonide, which instead induces opposite changes (e.g., increased mitochondrial oxidative phosphorylation). Finally, we provide evidence that budesonide inhibits PDAC growth, at least in part, through the tumor suppressor CDKN1C/p57Kip2. CONCLUSIONS: Collectively, our study reveals that the microenvironment influences the susceptibility of PDAC cells to GCs and provides unprecedented evidence for the anti-proliferative activity of budesonide on PDAC cells in 3D conditions, in vitro and in vivo. Our findings may explain, at least in part, the reason for the lower occurrence of pancreatic cancer in asthmatic patients and suggest a potential suitability of budesonide for clinical trials as a therapeutic approach to fight pancreatic cancer.


Assuntos
Budesonida , Proliferação de Células , Metabolismo Energético , Neoplasias Pancreáticas , Humanos , Budesonida/farmacologia , Budesonida/uso terapêutico , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Metabolismo Energético/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Movimento Celular/efeitos dos fármacos
2.
Pharmaceutics ; 15(7)2023 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-37514083

RESUMO

Small molecules that can modulate or stabilize cell-cell interactions are valuable tools for investigating the impact of collective cell behavior on various biological processes such as development/morphogenesis, tissue regeneration and cancer progression. Recently, we showed that budesonide, a glucocorticoid widely used as an anti-asthmatic drug, is a potent regulator of stem cell pluripotency. Here we tested the effect of different budesonide derivatives and identified CHD-030498 as a more effective analogue of budesonide. CHD-030498 was able to prevent stem cell pluripotency exit in different cell-based models, including embryonic stem-to-mesenchymal transition, spontaneous differentiation and 3D gastruloid development, and at lower doses compared to budesonide.

3.
Cells ; 11(14)2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35883568

RESUMO

In this paper, we summarize the current knowledge of the role of proline metabolism in the control of the identity of Embryonic Stem Cells (ESCs). An imbalance in proline metabolism shifts mouse ESCs toward a stable naïve-to-primed intermediate state of pluripotency. Proline-induced cells (PiCs), also named primitive ectoderm-like cells (EPLs), are phenotypically metastable, a trait linked to a rapid and reversible relocalization of E-cadherin from the plasma membrane to intracellular membrane compartments. The ESC-to-PiC transition relies on the activation of Erk and Tgfß/Activin signaling pathways and is associated with extensive remodeling of the transcriptome, metabolome and epigenome. PiCs maintain several properties of naïve pluripotency (teratoma formation, blastocyst colonization and 3D gastruloid development) and acquire a few traits of primed cells (flat-shaped colony morphology, aerobic glycolysis metabolism and competence for primordial germ cell fate). Overall, the molecular and phenotypic features of PiCs resemble those of an early-primed state of pluripotency, providing a robust model to study the role of metabolic perturbations in pluripotency and cell fate decisions.


Assuntos
Blastocisto , Células-Tronco Embrionárias , Animais , Blastocisto/metabolismo , Diferenciação Celular , Camundongos , Prolina/metabolismo , Transcriptoma
4.
Stem Cell Reports ; 17(11): 2548-2564, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36306780

RESUMO

3D embryonic stem cell (ESC) aggregates self-organize into embryo-like structures named gastruloids that recapitulate the axial organization of post-implantation embryos. Crucial in this process is the symmetry-breaking event that leads to the emergence of asymmetry and spatially ordered structures from homogeneous cell aggregates. Here, we show that budesonide, a glucocorticoid drug widely used to treat asthma, prevents ESC aggregates to break symmetry. Mechanistically, the effect of budesonide is glucocorticoid receptor independent. RNA sequencing and lineage fate analysis reveal that budesonide counteracts exit from pluripotency and modifies the expression of a large set of genes associated with cell migration, A-P axis formation, and WNT signaling. This correlates with reduced phenotypic and molecular cell heterogeneity, persistence of E-CADHERIN at the cell-cell interface, and cell aggregate compaction. Our findings reveal that cell-cell adhesion properties control symmetry breaking and cell fate transition in 3D gastruloids and suggest a potential adverse effect of budesonide on embryo development.


Assuntos
Embrião de Mamíferos , Células-Tronco Embrionárias , Adesão Celular , Células-Tronco Embrionárias/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Budesonida/farmacologia , Budesonida/metabolismo
5.
Stem Cell Reports ; 16(2): 354-369, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33482102

RESUMO

Floating spheroidal aggregates of mouse embryonic stem cells can develop into polarized/elongated organoids, namely gastruloids. We set up a high-performing assay to measure gastruloid formation efficiency (GFE), and found that GFE decreases as pluripotency progresses from naive (GFE ≥ 95%) to primed (GFE = 0) state. Specifically, we show that primed EpiSCs fail to generate proper cell aggregates, while early-primed EpiLCs aggregate but eventually fail to develop into elongated gastruloids. Moreover, we characterized proline-induced cells (PiCs), a LIF-dependent reversible early-primed state of pluripotency, and show that PiCs are able to generate gastruloids (GFE ∼ 50%) and are also competent to differentiate into primordial germ cell-like cells. Thus, we propose the GFE assay as a valuable functional tool to discriminate different states of the pluripotency continuum.


Assuntos
Desenvolvimento Embrionário , Fator de Crescimento Epidérmico/metabolismo , Camadas Germinativas/metabolismo , Glicoproteínas de Membrana/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas de Neoplasias/metabolismo , Organogênese , Organoides/embriologia , Células-Tronco Pluripotentes/metabolismo , Animais , Diferenciação Celular , Fator de Crescimento Epidérmico/genética , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética
6.
Front Cell Dev Biol ; 9: 728576, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34458276

RESUMO

Herein, we review the multifaceted roles of proline in cell biology. This peculiar cyclic imino acid is: (i) A main precursor of extracellular collagens (the most abundant human proteins), antimicrobial peptides (involved in innate immunity), salivary proteins (astringency, teeth health) and cornifins (skin permeability); (ii) an energy source for pathogenic bacteria, protozoan parasites, and metastatic cancer cells, which engage in extracellular-protein degradation to invade their host; (iii) an antistress molecule (an osmolyte and chemical chaperone) helpful against various potential harms (UV radiation, drought/salinity, heavy metals, reactive oxygen species); (iv) a neural metabotoxin associated with schizophrenia; (v) a modulator of cell signaling pathways such as the amino acid stress response and extracellular signal-related kinase pathway; (vi) an epigenetic modifier able to promote DNA and histone hypermethylation; (vii) an inducer of proliferation of stem and tumor cells; and (viii) a modulator of cell morphology and migration/invasiveness. We highlight how proline metabolism impacts beneficial tissue regeneration, but also contributes to the progression of devastating pathologies such as fibrosis and metastatic cancer.

7.
Cancer Res ; 79(13): 3235-3250, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31061065

RESUMO

Collagen prolyl hydroxylation (CPH), which is catalyzed by prolyl 4-hydroxylase (P4H), is the most prevalent posttranslational modification in humans and requires vitamin C (VitC). Here, we demonstrate that CPH acts as an epigenetic modulator of cell plasticity. Increased CPH induced global DNA/histone methylation in pluripotent stem and tumor cells and promoted cell state transition (CST). Interfering with CPH by either genetic ablation of P4H subunit alpha-2 (P4HA2) or pharmacologic treatment reverted epigenetic changes and antagonized CST. Mechanistically, we suggest that CPH modifies the epigenetic landscape by reducing VitC for DNA and histone demethylases. Repurposed drugs targeting CPH-mediated metabolic perturbation, such as the antiasthmatic budesonide, blocked metastatic dissemination of breast cancer cells in vivo by preventing mesenchymal transition. Our study provides mechanistic insights into how metabolic cues and epigenetic factors integrate to control CST and paves the way for the development of novel antimetastatic strategies. SIGNIFICANCE: A phenotype-based high-throughput screening reveals unforeseen metabolic control of cell plasticity and identifies budesonide as a drug candidate for metastatic cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/13/3235/F1.large.jpg.


Assuntos
Neoplasias da Mama/patologia , Colágeno/metabolismo , Epigênese Genética , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Células-Tronco Pluripotentes/patologia , Prolil Hidroxilases/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Hidroxilação , Células-Tronco Pluripotentes/metabolismo , Prolil Hidroxilases/genética
8.
Mol Cell Biol ; 23(12): 4401-15, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12773579

RESUMO

The AP-1 transcription factor plays an essential role in cell proliferation and tumorigenesis. It was previously shown that the fra-1 gene product is upregulated by various oncogenes and is involved in the in vitro and in vivo transformation of thyroid cells. Here we show that the ras oncogene-dependent accumulation of Fra-1 is mediated by a positive feedback mechanism which requires both transcriptional autoregulation and posttranslational stabilization of the protein. The oncogene-dependent transcriptional activation involves the cooperation between both Raf-dependent and Raf-independent pathways and is mediated by an AP-1 site within the fra-1 first intron, which becomes stably occupied by a transcriptionally active Fra-1-containing complex in ras-transformed cells. The posttranslational stabilization results in a drastic increase in the Fra-1 half-life in ras-transformed cells and is totally dependent on the activity of the MEK/ERK phosphorylation pathway. The analysis of the Fra-1 transactivation potential shows that the protein is able to stimulate a heterologous promoter in a ras-dependent manner, but the transactivating activity requires the recruitment of a heterodimeric partner. These data show that the alteration of multiple regulatory mechanisms is required for the constitutive activation of Fra-1 as a nuclear target of ras transformation.


Assuntos
Regulação da Expressão Gênica , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Transcrição Gênica , Proteínas ras/metabolismo , Animais , Northern Blotting , Linhagem Celular , Linhagem Celular Transformada , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cromatografia , DNA/metabolismo , Dimerização , Immunoblotting , Íntrons , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Modelos Genéticos , Mutação , Reação em Cadeia da Polimerase , Testes de Precipitina , Ligação Proteica , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Ativação Transcricional , Regulação para Cima
9.
Stem Cell Reports ; 8(1): 1-10, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28017658

RESUMO

Metabolites and cofactors are emerging as key regulators of cell plasticity and reprogramming, and their role in the control of pluripotency is just being discovered. Here we provide unprecedented evidence that embryonic stem cell (ESC) pluripotency relies on the relative levels of two physiological metabolites, namely ascorbic acid (vitamin C, VitC) and l-proline (l-Pro), which affect global DNA methylation, transcriptional profile, and energy metabolism. Specifically, while a high VitC/l-Pro ratio drives ESCs toward a naive state, the opposite condition (l-Pro excess) captures a fully reversible early primed pluripotent state, which depends on autocrine fibroblast growth factor and transforming growth factor ß signaling pathways. Our findings highlight the pivotal role of metabolites availability in controlling the pluripotency continuum from naive to primed states.


Assuntos
Ácido Ascórbico/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Prolina/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Autorrenovação Celular/efeitos dos fármacos , Análise por Conglomerados , Metilação de DNA/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Metaboloma , Metabolômica/métodos , Camundongos , MicroRNAs/genética , Células-Tronco Pluripotentes/citologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
10.
Mol Endocrinol ; 17(12): 2554-65, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14512522

RESUMO

Regulation of gene expression in haploid male germ cells follows a number of specific rules that differ from somatic cells. In this physiological context, transcriptional control mediated by the activator CREM (cAMP-responsive element modulator) represents an established paradigm. In somatic cells activation by CREM requires its phosphorylation at a unique regulatory site (Ser117) and subsequent interaction with the ubiquitous coactivator CBP (cAMP response element binding protein-binding protein). In testis, CREM transcriptional activity is controlled through interaction with a tissue-specific partner, ACT (activator of CREM in testis), which confers a powerful, phosphorylation-independent activation capacity. In addition to specialized transcription factors and coactivators, a variety of general factors of the basal transcriptional machinery, and their distinct tissue-specific isoforms, are highly expressed in testis, supporting the general notion that testis-specific gene expression requires specialized mechanisms. Here, we describe that CREM interacts with transcription factor IIA (TFIIA), a general transcription factor that stimulates RNA polymerase II-directed transcription. This association was identified by a two-hybrid screen, using a testis-derived cDNA library, and confirmed by coimmunoprecipitation. The interaction is restricted to the activator isoforms of CREM and does not require Ser117. Importantly, CREM does not interact with TFIIAtau-ALF, a testis-specific TFIIA homolog. CREM and TFIIA are expressed in a spatially and temporally coordinated fashion during the differentiation program of germ cells. The two proteins also colocalize intracellularly in spermatocyte and spermatid cells. These findings contribute to the understanding of the highly specialized rules of transcriptional regulation in haploid germ cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Repressoras , Espermatozoides/fisiologia , Fator de Transcrição TFIIA/metabolismo , Transcrição Gênica/genética , Animais , Clonagem Molecular , Modulador de Elemento de Resposta do AMP Cíclico , Regulação da Expressão Gênica/genética , Masculino , Camundongos , RNA/genética , RNA/isolamento & purificação , RNA Polimerase II/metabolismo , Saccharomyces cerevisiae , Ativação Transcricional
11.
Stem Cell Reports ; 1(4): 307-21, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24319666

RESUMO

Metabolites are emerging as key mediators of crosstalk between metabolic flux, cellular signaling, and epigenetic regulation of cell fate. We found that the nonessential amino acid L-proline (L-Pro) acts as a signaling molecule that promotes the conversion of embryonic stem cells into mesenchymal-like, spindle-shaped, highly motile, invasive pluripotent stem cells. This embryonic-stem-cell-to-mesenchymal-like transition (esMT) is accompanied by a genome-wide remodeling of the H3K9 and H3K36 methylation status. Consistently, L-Pro-induced esMT is fully reversible either after L-Pro withdrawal or by addition of ascorbic acid (vitamin C), which in turn reduces H3K9 and H3K36 methylation, promoting a mesenchymal-like-to-embryonic-stem-cell transition (MesT). These findings suggest that L-Pro, which is produced by proteolytic remodeling of the extracellular matrix, may act as a microenvironmental cue to control stem cell behavior.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Histonas/metabolismo , Prolina/farmacologia , Animais , Movimento Celular , Microambiente Celular , Citoesqueleto/ultraestrutura , Células-Tronco Embrionárias/citologia , Mesoderma/citologia , Metilação , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais , Transcriptoma
12.
Mol Biotechnol ; 50(3): 171-80, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21553282

RESUMO

The use of Embryonic Stem Cells (ESCs) holds considerable promise both for drug discovery programs and the treatment of degenerative disorders in regenerative medicine approaches. Nevertheless, the successful use of ESCs is still limited by the lack of efficient control of ESC self-renewal and differentiation capabilities. In this context, the possibility to modulate ESC biological properties and to obtain homogenous populations of correctly specified cells will help developing physiologically relevant screens, designed for the identification of stem cell modulators. Here, we developed a high throughput screening-suitable ESC neural differentiation assay by exploiting the Cell(maker) robotic platform and demonstrated that neural progenies can be generated from ESCs in complete automation, with high standards of accuracy and reliability. Moreover, we performed a pilot screening providing proof of concept that this assay allows the identification of regulators of ESC neural differentiation in full automation.


Assuntos
Automação Laboratorial/métodos , Diferenciação Celular , Ensaios de Triagem em Larga Escala/métodos , Células-Tronco Neurais/citologia , Animais , Linhagem Celular , Células-Tronco Embrionárias/citologia , Camundongos , Neurônios/citologia , Projetos Piloto , Reprodutibilidade dos Testes
13.
J Mol Cell Biol ; 3(2): 108-22, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21307025

RESUMO

The molecular mechanisms controlling mouse embryonic stem cell (ESC) metastability, i.e. their capacity to fluctuate between different states of pluripotency, are not fully resolved. We developed and used a novel automation platform, the Cell(maker), to screen a library of metabolites on two ESC-based phenotypic assays (i.e. proliferation and colony phenotype) and identified two metabolically related amino acids, namely l-proline (l-Pro) and l-ornithine (l-Orn), as key regulators of ESC metastability. Both compounds, but mainly l-Pro, force ESCs toward a novel epiblast stem cell (EpiSC)-like state, in a dose- and time-dependent manner. Unlike EpiSCs, l-Pro-induced cells (PiCs) contribute to chimeric embryos and rely on leukemia inhibitor factor (LIF) to self-renew. Furthermore, PiCs revert to ESCs or differentiate randomly upon removal of either l-Pro or LIF, respectively. Remarkably, PiC generation depends on both l-Pro metabolism (uptake and oxidation) and Fgf5 induction, and is strongly counteracted by antioxidants, mainly l-ascorbic acid (vitamin C, Vc). ESCs ↔ PiCs phenotypic transition thus represents a previously undefined dynamic equilibrium between pluripotent states, which can be unbalanced either toward an EpiSC-like or an ESC phenotype by l-Pro/l-Orn or Vc treatments, respectively. All together, our data provide evidence that ESC metastability can be regulated at a metabolic level.


Assuntos
Células-Tronco Embrionárias/metabolismo , Prolina/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Células Cultivadas , Células-Tronco Embrionárias/citologia , Feminino , Humanos , Masculino , Camundongos , Camundongos SCID , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
14.
Exp Cell Res ; 299(1): 209-26, 2004 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-15302588

RESUMO

Elucidating dynamic aspects of intracellular localization of proteins is essential to decipher their functional interaction networks. Although transcription factors lacking a detectable cytoplasmic fraction have been generally considered compartmentalized in the nucleus, some were found to shuttle into the cytoplasm, suggesting functional interactions therein. To further investigate how common, specific and quantitative is this traffic, we have employed the heterokaryon assay for a small-scale survey of nuclear factors not previously tested for their nucleo-cytoplasmic motion. We show that a subset of cAMP response element (CRE) binding proteins of the CREM type shuttles within a biologically meaningful time frame, revealing a continuous flow into the cytoplasm that persists during signaling. Their dynamic behavior, not involving the classical Exportin-1 pathway, could be ascribed to C-terminal sequences, containing, in addition to the bZIP domain and the NLS, a nuclear export activity and an inhibitory activity at an adjacent site. Other proteins examined in this study either did not shuttle significantly or, like CREB and distinct CREM isoforms, shuttled with markedly delayed kinetics, denoting considerable selectivity of this traffic. These findings raise the possibility that events associated with bi-directional transport and periodic transit through the cytoplasm may modulate activities of select nuclear transcription factors.


Assuntos
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Receptores Citoplasmáticos e Nucleares , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Núcleo Celular/genética , Cricetinae , Modulador de Elemento de Resposta do AMP Cíclico , Citoplasma/genética , Proteínas de Ligação a DNA/genética , Ácidos Graxos Insaturados/farmacologia , Células HeLa , Humanos , Carioferinas/efeitos dos fármacos , Carioferinas/genética , Carioferinas/metabolismo , Camundongos , Células NIH 3T3 , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína/genética , Proteínas Repressoras/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética , Proteína Exportina 1
15.
Proc Natl Acad Sci U S A ; 101(29): 10620-5, 2004 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-15247423

RESUMO

ACT [activator of cAMP-responsive element modulator (CREM) in testis] is a LIM-only protein that interacts with transcription factor CREM in postmeiotic male germ cells and enhances CREM-dependent transcription. CREM regulates many crucial genes required for spermatid maturation, and targeted mutation of the Crem gene in the mouse germ-line blocks spermatogenesis. Here we report the phenotype of mice in which targeted disruption of the act gene was obtained by homologous recombination. Whereas the seminiferous tubules of the act(-/-) mice contain all of the developmental stages of germ cells and the mice are fertile, the amount of mature sperm in the epididymis is drastically reduced. The residual sperm display severe abnormalities, including fully folded tails and aberrant head shapes. These results indicate that numerous postmeiotic genes under CREM control require the coactivator function of ACT. Thus, the fine-tuning of sperm development is achieved by the coordinated action of two transcriptional regulators.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Deleção de Genes , Proteínas Repressoras , Espermatogênese/fisiologia , Espermatozoides/anormalidades , Transativadores/metabolismo , Animais , Modulador de Elemento de Resposta do AMP Cíclico , Feminino , Fertilidade/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Cinesinas/metabolismo , Proteínas com Domínio LIM , Masculino , Camundongos , Camundongos Knockout , Proteínas Motores Moleculares/metabolismo , Fenótipo , Motilidade dos Espermatozoides , Espermatozoides/metabolismo , Espermatozoides/ultraestrutura , Testículo/citologia , Testículo/metabolismo , Transativadores/genética , Fatores de Transcrição , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA