Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int Immunol ; 21(7): 831-42, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19502585

RESUMO

B cell antigen receptor (BCR) cross-linking promotes proliferation and survival of mature B cells. Phosphoinositide-3-kinase-mediated down-regulation of pro-apoptotic and anti-mitogenic genes such as the Foxo family of transcription factors is an important component of this process. Previously, we demonstrated that BCR signaling decreases expression of transcripts for Foxo1, Foxo3 and Foxo4. We now show that BCR-induced down-regulation of Foxo3 and Foxo4 mRNA expression occurs via distinct mechanisms from those established for Foxo1. While Foxo1, Foxo3 and Foxo4 bind the same DNA sequence, the differential control of their expression upon B cell activation suggests that they may have unique functions in the B lineage. To begin to address this issue, we evaluated B cell development and function in Foxo3-/- mice. No effect of Foxo3 deficiency was observed with respect to the following parameters in the splenic B cell compartment: sub-population distribution, proliferation, in vitro differentiation and expression of the Foxo target genes cyclin G2 and B cell translocation gene 1. However, Foxo3-/- mice demonstrated increased basal levels of IgG2a, IgG3 and IgA. A significant reduction in pre-B cell numbers was also observed in Foxo3-/- bone marrow. Finally, recirculating B cells in the bone marrow and peripheral blood were decreased in Foxo3-/- mice, perhaps due to lower than normal expression of receptor for sphingosine-1 phosphate, which mediates egress from lymphoid organs. Thus, Foxo3 makes a unique contribution to B cell development, B cell localization and control of Ig levels.


Assuntos
Subpopulações de Linfócitos B/imunologia , Fatores de Transcrição Forkhead/metabolismo , Células Precursoras de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Anticorpos Anti-Idiotípicos/farmacologia , Subpopulações de Linfócitos B/efeitos dos fármacos , Butadienos/farmacologia , Proteínas de Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Cromonas/farmacologia , Ciclina G1 , Ciclina G2 , Ciclinas/imunologia , Ciclinas/metabolismo , Ciclosporina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Inibidores Enzimáticos/farmacologia , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Interleucina-7/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfolinas/farmacologia , Nitrilas/farmacologia , Fosfatidilinositol 3-Quinases/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Células Precursoras de Linfócitos B/efeitos dos fármacos , Receptores de Antígenos de Linfócitos B/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
2.
Circ Res ; 102(9): 1075-81, 2008 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-18356544

RESUMO

Stem and progenitor cell populations occupy a specialized niche and are consequently exposed to hypoxic as well as oxidative stresses. We have previously established that the multidrug resistance protein Abcg2 is the molecular determinant of the side population (SP) progenitor cell population. We observed that the cardiac SP cells increase in number more than 3-fold within 3 days of injury. Transcriptome analysis of the SP cells isolated from the injured adult murine heart reveals increased expression of cytoprotective transcripts. Overexpression of Abcg2 results in an increased ability to consume hydrogen peroxide and is associated with increased levels of alpha-glutathione reductase protein expression. Importantly, overexpression of Abcg2 also conferred a cell survival benefit following exposure to hydrogen peroxide. To further examine the molecular regulation of the Abcg2 gene, we demonstrated that hypoxia-inducible factor (HIF)-2alpha binds an evolutionary conserved HIF-2alpha response element in the murine Abcg2 promoter. Transcriptional assays reveal a dose-dependent activation of Abcg2 expression by HIF-2alpha. These results support the hypothesis that Abcg2 is a direct downstream target of HIF-2alpha which functions with other factors to initiate a cytoprotective program for this progenitor SP cell population that resides in the adult heart.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Células-Tronco Adultas/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Citoproteção , Células-Tronco Embrionárias/metabolismo , Traumatismos Cardíacos/prevenção & controle , Ativação Transcricional , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Células-Tronco Adultas/enzimologia , Células-Tronco Adultas/patologia , Animais , Linhagem Celular , Proliferação de Células , Células Cultivadas , Temperatura Baixa/efeitos adversos , Citoproteção/genética , Modelos Animais de Doenças , Células-Tronco Embrionárias/patologia , Perfilação da Expressão Gênica/métodos , Glutationa Redutase/metabolismo , Traumatismos Cardíacos/etiologia , Traumatismos Cardíacos/genética , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Peróxido de Hidrogênio/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção , Regulação para Cima
3.
Dev Biol ; 321(1): 197-204, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18601916

RESUMO

In mammals, oocytes are packaged into compact structures-primordial follicles-which remain inert for prolonged intervals until individual follicles resume growth via a process known as primordial follicle activation. Here we show that the phosphoinositide 3-kinase (PI3K) signalling pathway controls primordial follicle activation through the forkhead transcription factor Foxo3. Within oocytes, Foxo3 is regulated by nucleocytoplasmic shuttling. Foxo3 is imported into the nucleus during primordial follicle assembly, and is exported upon activation. Oocyte-specific ablation of Pten resulted in PI3K-induced Akt activation, Foxo3 hyperphosphorylation, and Foxo3 nuclear export, thereby triggering primordial follicle activation, defining the steps by which the PI3K pathway and Foxo3 control this process. Inducible ablation of Pten and Foxo3 in adult oocytes using a new tool for genetic analysis of the germline, Vasa-Cre(ERT2), showed that this pathway functions throughout life. Thus, a principal physiologic role of the PI3K pathway is to control primordial follicle activation via Foxo3.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Oócitos/crescimento & desenvolvimento , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Proteína Forkhead Box O3 , Camundongos , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais
4.
Genetics ; 177(1): 179-94, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17660561

RESUMO

Female infertility syndromes are among the most prevalent chronic health disorders in women, but their genetic basis remains unknown because of uncertainty regarding the number and identity of ovarian factors controlling the assembly, preservation, and maturation of ovarian follicles. To systematically discover ovarian fertility genes en masse, we employed a mouse model (Foxo3) in which follicles are assembled normally but then undergo synchronous activation. We developed a microarray-based approach for the systematic discovery of tissue-specific genes and, by applying it to Foxo3 ovaries and other samples, defined a surprisingly large set of ovarian factors (n = 348, approximately 1% of the mouse genome). This set included the vast majority of known ovarian factors, 44% of which when mutated produce female sterility phenotypes, but most were novel. Comparative profiling of other tissues, including microdissected oocytes and somatic cells, revealed distinct gene classes and provided new insights into oogenesis and ovarian function, demonstrating the utility of our approach for tissue-specific gene discovery. This study will thus facilitate comprehensive analyses of follicle development, ovarian function, and female infertility.


Assuntos
Fatores de Transcrição Forkhead/fisiologia , Genes/fisiologia , Genoma , Infertilidade Feminina/metabolismo , Folículo Ovariano/fisiologia , Animais , Northern Blotting , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Hibridização In Situ , Infertilidade Feminina/patologia , Lasers , Meiose , Ciclo Menstrual , Camundongos , Camundongos Knockout , Microdissecção , Análise de Sequência com Séries de Oligonucleotídeos , Oócitos/fisiologia , Oócitos/ultraestrutura , Oogênese/fisiologia , Folículo Ovariano/ultraestrutura , Sondas RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Hum Reprod ; 23(1): 216-21, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17959613

RESUMO

BACKGROUND: The forkhead transcription factor Foxo3 is a master regulator and potent suppressor of primordial follicle activation. Loss of Foxo3 function in the mouse leads to premature ovarian failure (POF) due to global follicle activation. METHODS AND RESULTS: Here, we show that the mouse Foxo3 locus is haploinsufficient, and that Foxo3-/+ females undergo early reproductive senescence consistent with an increased rate of primordial follicle utilization. Then, to determine if heterozygous or homozygous polymorphisms or mutations of the human orthologue FOXO3 contribute to POF or idiopathic primary amenorrhea (PA), we sequenced the exons and flanking splice sequences of the gene in a large number of women with idiopathic POF (n = 273) or PA (n = 29). A total of eight single-nucleotide polymorphisms (SNPs) were identified, revealing a substantial amount of genetic variation at this locus. Allelic frequencies in control samples excluded several of these variants as causal. For the remaining variants, site-directed mutagenesis was performed to assess their functional impact. However, these rare sequence variants were not associated with significant decreases in FOXO3 activity. CONCLUSIONS: Taken together, our findings suggest that, despite the potential for FOXO3 haploinsufficiency to cause ovarian failure, FOXO3 mutations or common SNPs are not a common cause of either POF or PA.


Assuntos
Amenorreia/genética , Fatores de Transcrição Forkhead/genética , Variação Genética , Insuficiência Ovariana Primária/genética , Adulto , Animais , Sequência de Bases , Éxons , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Frequência do Gene , Haplótipos , Heterozigoto , Humanos , Masculino , Camundongos , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase/métodos , Polimorfismo de Nucleotídeo Único
6.
Med Clin (Barc) ; 129(8): 292-4, 2007 Sep 08.
Artigo em Espanhol | MEDLINE | ID: mdl-17878022

RESUMO

BACKGROUND AND OBJECTIVE: Women with polycystic ovary syndrome (PCOS) exhibit frequently risk factors that predispose to cardiovascular disease. Hyperhomocysteinemia is an independent risk factor for this disease. The aim of this study was to know whether young women with PCOS have increased homocysteine levels. We also analyzed their possible relation with folate and vitamin B12 levels. PATIENTS AND METHOD: Thirty nine patients with PCOS were studied; (age: mean [standard deviation] 28.9 [5.8] years), and 39 healthy women similar in age. We evaluated in all of them: smoking, menstrual cycles, hirsutism, body mass index, metabolic syndrome and levels of homocysteine, lipids, glucose, creatinine, folate, vitamin B12, follicle-stimulating hormone (FSH), luteinizing hormone (LH) and androstendione. RESULTS: Menstrual cycles, hirsutism, androstendione, LH levels and LH/FSH were higher, as we expected, in patients with PCOS. Moreover, patients had increased homocysteine (9.1 [2.1] vs 6.4 [1.8] micromol/L; p < 0.001) and glucose levels (99 [13] vs 88 [10] mg/dl; p < 0.001), a higher frequency of abnormal fasting glycemia (> 110 mg/dl) (23% vs 2.5%; p =.01) and lower folate levels (7.6 [3.7] vs 10.2 [3.6] ng/ml; p = 0.02). A multiple linear regression showed a negative association between homocysteine and folate levels (r2 = 0.05; p =.02). CONCLUSIONS: Homocysteinemia is increased in women with PCOS, and it is negatively associated with folate levels.


Assuntos
Hiper-Homocisteinemia/epidemiologia , Síndrome do Ovário Policístico/epidemiologia , Adulto , Biomarcadores , Feminino , Hormônio Foliculoestimulante/sangue , Humanos , Hiper-Homocisteinemia/sangue , Hormônio Luteinizante/sangue , Síndrome do Ovário Policístico/sangue
7.
Circ Res ; 95(4): 389-97, 2004 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-15242968

RESUMO

Transcriptome-wide analysis of dynamically regulated progenitor cell populations has the potential to elucidate key aspects of cardiac development. The heart, as the first organ to develop in the mammal, is a technically challenging but clinically relevant target for study. To define the transcriptional program of the cardiac progenitor, we used a novel transgenic strategy and fluorescence-activated cell sorting to reliably label and isolate cardiac progenitors directly from mouse embryos. Pure populations of cardiac progenitor cells were isolated from the cardiac crescent and 2 subsequent stages of heart development: the linear heart tube and the looping heart. RNA was isolated from stage-specific cardiac progenitors and subjected to transcriptome analysis by oligonucleotide array hybridization. The cardiac transcriptional regulatory programs were compared with the molecular programs of age-matched noncardiac embryonic cells, embryonic stem cells, adult cardiomyocytes, and each other to identify sets of genes exhibiting differential expression in the cardiac progenitor cell population. These results define the transcriptional profile of mammalian cardiac progenitor cells and provide insight into the molecular regulation of the earliest periods of heart development.


Assuntos
Regulação da Expressão Gênica , Coração/embriologia , Miócitos Cardíacos/metabolismo , Células-Tronco/metabolismo , Transcrição Gênica , Animais , Proteínas de Bactérias/análise , Proteínas de Bactérias/genética , Cruzamentos Genéticos , Citometria de Fluxo , Perfilação da Expressão Gênica , Genes Reporter , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfogênese/genética , Miócitos Cardíacos/citologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia
8.
Physiol Genomics ; 14(3): 261-71, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12799472

RESUMO

Muscle regeneration is a complex process requiring the coordinated interaction between the myogenic progenitor cells or satellite cells, growth factors, cytokines, inflammatory components, vascular components and the extracellular matrix (ECM). Previous studies have elegantly described the physiological modulation of the regenerative process in response to muscle injury, but the molecular response that characterizes stages of the repair process remains ill-defined. The recent completion of the Human and Mouse Genome Projects and the advent of technologies such as high-density oligonucleotide array analysis facilitate an expanded analysis of complex processes such as muscle regeneration. In the present study, we define cellular and molecular events that characterize stages of muscle injury and regeneration. Utilization of transcriptional profiling strategies revealed coordinated expression of growth factors [i.e., Tgfb1, Igf1, Egf, chemokine (C-C motif) ligand 6 and 7], the fetal myogenic program (Myod1, Myf5, Myf6), and the biomatrix (procollagen genes, Mmp3, Mmp9, biglycan, periostin) during muscle regeneration. Corroboration of the transcriptional profiling analysis included quantitative real-time RT-PCR and in situ hybridization analyses of selected candidate genes. In situ hybridization studies for periostin [osteoblast-specific factor 2 (fasciclin I-like)] and biglycan revealed that these genes are restricted to mesenchymal derivatives during embryogenesis and are significantly regulated during regeneration of the injured hindlimb skeletal muscle. We conclude that muscle regeneration is a complex process that requires the coordinated modulation of the inflammatory response, myogenic progenitor cells, growth factors, and ECM for complete restoration of muscle architecture.


Assuntos
Matriz Extracelular/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Músculo Esquelético/fisiopatologia , Análise de Sequência com Séries de Oligonucleotídeos , Regeneração/genética , Células 3T3 , Animais , Biglicano , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/fisiologia , Linhagem Celular , Matriz Extracelular/fisiologia , Proteínas da Matriz Extracelular , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Membro Posterior , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/embriologia , Músculo Esquelético/patologia , Mioblastos/química , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Mioblastos/patologia , Proteoglicanas/biossíntese , Proteoglicanas/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta/fisiologia , Fator de Crescimento Transformador beta1
9.
Physiol Genomics ; 12(1): 53-60, 2002 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-12502795

RESUMO

The atria and ventricles of the heart have distinct development, structure, and physiology. However, only a few of the genes that underlie the differences between these tissues are known. We used a murine cardiac cDNA microarray to identify genes differentially expressed in the atria and ventricles. The reliability of these findings is supported by highly concordant repetition of hybridization, recognition of previously known atrial and ventricular isoforms of contractile proteins, and confirmation of results by quantitative PCR and in situ hybridization. We examined the most differentially regulated genes for evolutionarily conserved noncoding sequences and found that atrial-expressed genes have more predicted myocyte enhancer factor-2 (MEF2) binding sites than ventricle-predominant genes. We confirmed that messages for MEF2 family members are more abundant in the atria, as are their protein products. Moreover, the activity of a transgenic reporter construct for MEF2 activity is preferentially upregulated in the atria in response to hypertrophic stimuli. This study provides a greater understanding of the molecular differences between atria and ventricles and establishes the framework for an anatomically detailed evaluation of cardiac transcriptional regulation.


Assuntos
Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Transcrição Gênica , Animais , Sítios de Ligação , Sequência Conservada , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Feminino , Perfilação da Expressão Gênica , Genômica , Coração/anatomia & histologia , Hibridização In Situ , Fatores de Transcrição MEF2 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Regulação Miogênica , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Oncol Res ; 14(3): 147-54, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14760863

RESUMO

In this study we evaluated a mono-tetrahydrofuranic subgroup of natural acetogenins that had shown in previous enzyme inhibition studies different potency trends compared with the bis-tetrahydrofuranic acetogenin subgroup. The compounds were tested against colon, breast, lung, liver, and ovarian tumor cell lines. A drug-resistant ovarian cell line was also included in the panel. In general the compounds were more potent than doxorubicin. The goal was to determine how well the mitochondrial complex I inhibition correlates with the in vitro antitumor potency of these natural mono-tetrahydrofuranic acetogenins and of some derivatives. The results indicate that both the reduction of the terminal gamma-lactone after its translactonization and the introduction of an hydroxylimine group in the alkyl chain, near the mono-tetrahydrofuranic moiety, increased the antitumor activity, even against the doxorubicin-resistant cell line.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Álcoois Graxos/química , Álcoois Graxos/farmacologia , Furanos/química , Lactonas/química , Lactonas/farmacologia , Acetogeninas , Linhagem Celular Tumoral , Complexo I de Transporte de Elétrons/metabolismo , Furanos/síntese química , Furanos/farmacologia , Humanos , Concentração Inibidora 50 , Lactonas/síntese química , Estrutura Molecular , Complexos Multienzimáticos/metabolismo , NADH NADPH Oxirredutases/metabolismo , Relação Estrutura-Atividade , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia
11.
J Clin Invest ; 121(9): 3456-66, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21865646

RESUMO

Spermatogonial stem cells (SSCs) capable of self-renewal and differentiation are the foundation for spermatogenesis. Although several factors important for these processes have been identified, the fundamental mechanisms regulating SSC self-renewal and differentiation remain unknown. Here, we investigated a role for the Foxo transcription factors in mouse spermatogenesis and found that Foxo1 specifically marks mouse gonocytes and a subset of spermatogonia with stem cell potential. Genetic analyses showed that Foxo1 was required for both SSC homeostasis and the initiation of spermatogenesis. Combined deficiency of Foxo1, Foxo3, and Foxo4 resulted in a severe impairment of SSC self-renewal and a complete block of differentiation, indicating that Foxo3 and Foxo4, although dispensable for male fertility, contribute to SSC function. By conditional inactivation of 3-phosphoinositide-dependent protein kinase 1 (Pdk1) and phosphatase and tensin homolog (Pten) in the male germ line, we found that PI3K signaling regulates Foxo1 stability and subcellular localization, revealing that the Foxos are pivotal effectors of PI3K-Akt signaling in SSCs. We also identified a network of Foxo gene targets--most notably Ret--that rationalized the maintenance of SSCs by the Foxos. These studies demonstrate that Foxo1 expression in the spermatogenic lineage is intimately associated with the stem cell state and revealed what we believe to be novel Foxo-dependent mechanisms underlying SSC self-renewal and differentiation, with implications for common diseases, including male infertility and testicular cancer, due to abnormalities in SSC function.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Espermatogênese/fisiologia , Espermatogônias/citologia , Células-Tronco/fisiologia , Animais , Proteínas de Ciclo Celular , Diferenciação Celular/fisiologia , Feminino , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Infertilidade Masculina/fisiopatologia , Masculino , Camundongos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil , Transdução de Sinais/fisiologia , Espermatogônias/fisiologia , Células-Tronco/citologia , Testículo/citologia
12.
Dis Model Mech ; 3(3-4): 181-93, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20142330

RESUMO

Endometrial cancer--the most common malignancy of the female reproductive tract--arises from the specialized epithelial cells that line the inner surface of the uterus. Although significant advances have been made in our understanding of this disease in recent years, one significant limitation has been the lack of a diverse genetic toolkit for the generation of mouse models. We identified a novel endometrial-specific gene, Sprr2f, and developed a Sprr2f-Cre transgene for conditional gene targeting within endometrial epithelium. We then used this tool to generate a completely penetrant Lkb1 (also known as Stk11)-based mouse model of invasive endometrial cancer. Strikingly, female mice with homozygous endometrial Lkb1 inactivation did not harbor discrete endometrial neoplasms, but instead underwent diffuse malignant transformation of their entire endometrium with rapid extrauterine spread and death, suggesting that Lkb1 inactivation was sufficient to promote the development of invasive endometrial cancer. Mice with heterozygous endometrial Lkb1 inactivation only rarely developed tumors, which were focal and arose with much longer latency, arguing against the idea--suggested by some prior studies--that Lkb1 is a haploinsufficient tumor suppressor. Lastly, the finding that endometrial cancer cell lines were especially sensitive to the mTOR (mammalian target of rapamycin) inhibitor rapamycin prompted us to test its efficacy against Lkb1-driven endometrial cancers. Rapamycin monotherapy not only greatly slowed disease progression, but also led to striking regression of pre-existing tumors. These studies demonstrate that Lkb1 is a uniquely potent endometrial tumor suppressor, but also suggest that the clinical responses of some types of invasive cancers to mTOR inhibitors may be linked to Lkb1 status.


Assuntos
Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Sirolimo/uso terapêutico , Proteínas Quinases Ativadas por AMP , Animais , Linhagem Celular Tumoral , Clonagem Molecular , Proteínas Ricas em Prolina do Estrato Córneo/genética , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais , Neoplasias do Endométrio/patologia , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Endométrio/patologia , Ativação Enzimática/efeitos dos fármacos , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Epitélio/patologia , Feminino , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica , Especificidade de Órgãos/efeitos dos fármacos , Penetrância , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/deficiência , Elementos de Resposta/genética , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
13.
PLoS One ; 4(4): e5137, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19340305

RESUMO

Human Papilloma Virus (HPV) is the etiologic agent for cervical cancer. Yet, infection with HPV is not sufficient to cause cervical cancer, because most infected women develop transient epithelial dysplasias that spontaneously regress. Progression to invasive cancer has been attributed to diverse host factors such as immune or hormonal status, as no recurrent genetic alterations have been identified in cervical cancers. Thus, the pressing question as to the biological basis of cervical cancer progression has remained unresolved, hampering the development of novel therapies and prognostic tests. Here we show that at least 20% of cervical cancers harbor somatically-acquired mutations in the LKB1 tumor suppressor. Approximately one-half of tumors with mutations harbored single nucleotide substitutions or microdeletions identifiable by exon sequencing, while the other half harbored larger monoallelic or biallelic deletions detectable by multiplex ligation probe amplification (MLPA). Biallelic mutations were identified in most cervical cancer cell lines; HeLa, the first human cell line, harbors a homozygous 25 kb deletion that occurred in vivo. LKB1 inactivation in primary tumors was associated with accelerated disease progression. Median survival was only 13 months for patients with LKB1-deficient tumors, but >100 months for patients with LKB1-wild type tumors (P = 0.015, log rank test; hazard ratio = 0.25, 95% CI = 0.083 to 0.77). LKB1 is thus a major cervical tumor suppressor, demonstrating that acquired genetic alterations drive progression of HPV-induced dysplasias to invasive, lethal cancers. Furthermore, LKB1 status can be exploited clinically to predict disease recurrence.


Assuntos
Mutação , Proteínas Serina-Treonina Quinases/genética , Neoplasias do Colo do Útero/patologia , Quinases Proteína-Quinases Ativadas por AMP , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Progressão da Doença , Feminino , Deleção de Genes , Células HeLa , Humanos , Pessoa de Meia-Idade , Neoplasias do Colo do Útero/genética
14.
Genesis ; 45(6): 413-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17551945

RESUMO

Cell type-specific genetic modification using the Cre/loxP system is a powerful tool for genetic analysis of distinct cell lineages. Because of the exquisite specificity of Vasa expression (confined to the germ cell lineage in invertebrate and vertebrate species), we hypothesized that a Vasa promoter-driven transgenic Cre line would prove useful for the germ cell lineage-specific inactivation of genes. Here we describe a transgenic mouse line, Vasa-Cre, where Cre is efficiently and specifically expressed in germ cells. Northern analysis showed that transgene expression was confined to the gonads. Cre-mediated recombination with the Rosa26-lacZ reporter was observed beginning at approximately e15, and was >95% efficient in male and female germ cells by birth. Although there was a potent maternal effect with some animals showing more widespread recombination, there was no ectopic activity in most adults. This Vasa-Cre transgenic line should thus prove useful for genetic analysis of diverse aspects of gametogenesis and as a general deletor line.


Assuntos
RNA Helicases DEAD-box/genética , Gametogênese/genética , Células Germinativas/enzimologia , Integrases/genética , Camundongos Transgênicos/genética , Animais , Genótipo , Camundongos , Regiões Promotoras Genéticas , beta-Galactosidase/análise , beta-Galactosidase/genética
15.
Reproduction ; 133(5): 855-63, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17616716

RESUMO

Primordial follicles are long-lived structures assembled early in life. The mechanisms that control the balance between the conservation and the activation of primordial follicles are critically important for fertility and dictate the onset of menopause. The forkhead transcription factor Foxo3 serves an essential role in these processes by suppressing the growth of primordial follicles, thereby preserving them until later in life. While other factors regulating primordial follicle growth have been described, most serve multiple functions at several stages of female germ cell or follicle development, and corresponding mouse mutants exhibit pleiotropic phenotypes with disruption of multiple stages of follicle assembly, development, or survival. To investigate the possibility that Foxo3 also functions in other aspects of ovarian development beyond its known role in primordial follicle activation (PFA), we performed detailed analyses of mouse ovaries including electron microscopy to study primordial follicle structure, assembly, and early growth. These analyses revealed that the timing of primordial follicle assembly, early oocyte survival, and the expression of early germ line markers were unaffected in early Foxo3 ovaries. Taken together, these studies demonstrate that the phenotype associated with Foxo3 deficiency is remarkably specific for PFA and further support the placement of Foxo3 in a unique phenotypic class among mammalian female sterile mutants. Lastly, we discuss the implications of the specificity of this mutant phenotype with regard to the hypothesis that oocyte regeneration may occur in adults and serves as a means to replenish oocytes lost via natural physiological processes.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Infertilidade Feminina/metabolismo , Folículo Ovariano/fisiologia , Animais , Apoptose , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/análise , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Infertilidade Feminina/patologia , Ciclo Menstrual , Camundongos , Camundongos Knockout , Oócitos/fisiologia , Oócitos/ultraestrutura , Folículo Ovariano/ultraestrutura
16.
Dev Biol ; 288(2): 502-13, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16310178

RESUMO

The Serum Response Factor (SRF) coactivator myocardin stimulates the transcription of multiple muscle genes during cardiac and smooth muscle development. Mouse embryos lacking myocardin die during the earliest stages of smooth muscle development and fail to express multiple smooth muscle marker genes in the embryonic dorsal aorta and other vascular structures. In this study, we used mutant embryonic stem cell lines to further define the role of myocardin in smooth muscle differentiation and vascular development. Misexpression of myocardin in undifferentiated muscle stem cells resulted in efficient activation of smooth muscle genes, and weaker activation of genes involved in cardiac and skeletal muscle differentiation. Remarkably, myocardin(-/-) embryonic stem cell lines differentiated into smooth muscle cells in vitro, although these cells expressed significantly decreased levels of smooth muscle contractile genes. Moreover, genetically labeled myocardin(-/-) ES cells were able to contribute to smooth muscle lineages in vivo. These results indicate that while myocardin function is sufficient for activation of SRF-dependent muscle gene expression in multiple cell types, myocardin-independent mechanism(s) can suffice for expression in some smooth muscle lineages.


Assuntos
Diferenciação Celular/fisiologia , Perfilação da Expressão Gênica , Músculo Liso/citologia , Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Células-Tronco/citologia , Transativadores/metabolismo , Animais , Linhagem Celular , Linhagem da Célula/fisiologia , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Camundongos Mutantes , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Músculo Liso/embriologia , Mutação , Miocárdio/citologia , Miocárdio/metabolismo , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Transativadores/genética
17.
Genesis ; 37(2): 57-63, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14595841

RESUMO

As stem cells constitute a rare cell pool, a global analysis of gene expression is typically not possible. Here, we use a T7-based RNA amplification method combined with high-density oligonucleotide array technology to analyze gene expression. We isolated RNA from ES cells (100-100,000 cells), subjected them to two rounds of amplification, and compared each sample using microarray analysis. RNA isolation and amplification was highly reproducible and sensitive for the analysis of low abundant transcripts. Greater than 93% of the transcripts expressed were changed less than 2-fold when comparing the results of amplified RNA (100 cells to 100,000 cells) to unamplified RNA (isolated from 1,000,000 cells). This transcriptional analysis resulted in minimal skewing of gene expression. Using this technology, we analyzed the genetic programs of ES cells, STO feeder cells, and adult cardiomyocytes. We conclude that these technologies can be applied to the analysis of the genetic programs of rare cell populations and ultimately this analysis will enhance our understanding of the regulatory mechanisms of stem cell populations.


Assuntos
Perfilação da Expressão Gênica/métodos , Expressão Gênica , Técnicas de Amplificação de Ácido Nucleico/métodos , Células-Tronco/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular , Embrião de Mamíferos/citologia , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA/isolamento & purificação , Reprodutibilidade dos Testes , Células-Tronco/citologia
18.
J Muscle Res Cell Motil ; 24(8): 587-92, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14870974

RESUMO

Vertebrate skeletal myofibers are heterogeneous with respect to their metabolic, contractile and morphological properties. To better understand skeletal myofiber diversity and plasticity at the transcriptional level, we carried out a whole-genome gene expression analysis of skeletal muscles composed primarily of slow/oxidative fibers (soleus) and fast fibers (extensor digitorum longus, EDL). We also followed gene expression changes in plantaris muscles from mice undergoing voluntary wheel running, a protocol that triggers transformation of glycolytic fibers into oxidative ones. Microarray analysis identified 70 genes differentially expressed by 3-fold or greater in soleus vs. EDL muscles and 15 genes up-regulated in exercised vs. sedentary plantaris muscles. A subset of these results were verified by northern blot and/or real-time RT-PCR analyses. Our results expand knowledge of the differences among various types of skeletal myofibers and their adaptation to exercise at the transcriptional level.


Assuntos
Fibras Musculares de Contração Rápida/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares de Contração Lenta/metabolismo , Condicionamento Físico Animal , Resistência Física , Animais , Northern Blotting , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Músculo Esquelético/metabolismo
19.
Science ; 296(5566): 349-52, 2002 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-11951046

RESUMO

Endurance exercise training promotes mitochondrial biogenesis in skeletal muscle and enhances muscle oxidative capacity, but the signaling mechanisms involved are poorly understood. To investigate this adaptive process, we generated transgenic mice that selectively express in skeletal muscle a constitutively active form of calcium/calmodulin-dependent protein kinase IV (CaMKIV*). Skeletal muscles from these mice showed augmented mitochondrial DNA replication and mitochondrial biogenesis, up-regulation of mitochondrial enzymes involved in fatty acid metabolism and electron transport, and reduced susceptibility to fatigue during repetitive contractions. CaMK induced expression of peroxisome proliferator-activated receptor gamma coactivator 1 (PGC-1), a master regulator of mitochondrial biogenesis in vivo, and activated the PGC-1 gene promoter in cultured myocytes. Thus, a calcium-regulated signaling pathway controls mitochondrial biogenesis in mammalian cells.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Animais , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Replicação do DNA , DNA Mitocondrial/biossíntese , Transporte de Elétrons , Ácidos Graxos/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias Musculares/enzimologia , Contração Muscular , Fadiga Muscular , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/enzimologia , Músculo Esquelético/ultraestrutura , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transgenes , Regulação para Cima
20.
Stem Cells ; 22(7): 1305-20, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15579648

RESUMO

Muscle progenitor cells (satellite cells) function in the maintenance and repair of adult skeletal muscle. Side population (SP) cells are enriched in repopulating activity and also reside in adult skeletal muscle. In this study, we observed that Abcg2 is a determinant of the SP cell phenotype. Using reverse transcription polymerase chain reaction and immunohistochemical techniques, we localized Abcg2-expressing cells in the interstitium and in close approximation to the vasculature of adult skeletal muscle. Muscle SP cells are able to differentiate into myotubes and increase in number after cardiotoxin-induced muscle injury. Similar to myogenic progenitor cells, muscle SP cells express Foxk1 and are decreased in number in Foxk1 mutant skeletal muscle. Using emerging technologies, we examine the molecular signature of muscle SP cells from normal, injured, and Foxk1 mutant skeletal muscle to define common and distinct molecular programs. We propose that muscle SP cells are progenitor cells that participate in repair and regeneration of adult skeletal muscle.


Assuntos
Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/fisiologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/fisiologia , Animais , Diferenciação Celular , Separação Celular , Proteínas Cardiotóxicas de Elapídeos/farmacologia , Citometria de Fluxo , Fatores de Transcrição Forkhead , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculos/patologia , Distrofias Musculares/patologia , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares/genética , Hibridização de Ácido Nucleico , Fenótipo , RNA/metabolismo , Regeneração , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Fatores de Tempo , Fatores de Transcrição/genética , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa