Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Funct Integr Genomics ; 15(3): 271-5, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25403593

RESUMO

MicroRNAs (miRNAs) are known to influence ovarian cell proliferation, apoptosis and hormone release, but it remains unknown whether miRNAs affect ovarian functions via transcription factors. We examined the effect of miRNAs on nuclear factor-κappaB (NF-kB) (p65) expression in human ovarian luteinized granulosa cells. We transfected cultured primary human ovarian luteinized granulosa cells with 80 different constructs encoding human pre-miRNAs and then evaluated NF-kB (p65) expression (percentage of cells containing p65) by immunocytochemistry. We found that 21 of the constructs stimulated NF-kB (p65) expression and 18 of the constructs inhibited NF-kB (p65) expression. This is the first direct demonstration that miRNAs affect NF-kB (p65) expression and the first genome-scale miRNA screen to identify upregulation and downregulation of NF-kB accumulation by miRNAs in the ovary. Novel miRNAs that affect the NF-kB signalling pathway could be useful for the control of NF-kB-dependent reproductive processes and the treatment of NF-kB-dependent reproductive disorders.


Assuntos
Células da Granulosa/metabolismo , MicroRNAs/metabolismo , Fator de Transcrição RelA/metabolismo , Adulto , Sobrevivência Celular , Células Cultivadas , Feminino , Humanos , MicroRNAs/genética , Transfecção
2.
Blood ; 118(7): 1899-902, 2011 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-21685371

RESUMO

MicroRNAs (miRNAs) are small, noncoding RNA molecules that regulate growth and differentiation. miRNAs are frequently located at cancer-specific fragile sites in the human genome, such as chromosome 7q. The nuclear oncogene SKI is up-regulated in acute myeloid leukemia (AML) with -7/del7q. Here we asked whether loss of miRNAs on chromosome 7q may explain this up-regulation. miR-29a expression was found to be down-regulated in AML with -7/del7q. Forced expression of miR-29a down-regulated Ski and its target gene, Nr-CAM, whereas miR-29a inhibition induced Ski expression. Luciferase assays validated a functional binding site for miR-29a in the 3' untranslated region of SKI. Finally, in samples of AML patients, we observed an inverse correlation of Ski and miR-29a expression, respectively. In conclusion, up-regulation of Ski in AML with -7/del7q is caused by loss of miR-29a. miR-29a may therefore function as an important tumor suppressor in AML by restraining expression of the SKI oncogene.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/genética , MicroRNAs/genética , Proteínas Proto-Oncogênicas/genética , Regiões 3' não Traduzidas , Linhagem Celular Tumoral , Cromossomos Humanos Par 7 , Regulação para Baixo , Humanos , Regulação para Cima
3.
J Cell Physiol ; 223(1): 49-56, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20039279

RESUMO

Previous studies have shown that microRNAs (miRNAs) can control steroidogenesis in cultured granulosa cells. In this study we wanted to determine if miRNAs can also affect proliferation and apoptosis in human ovarian cells. The effect of transfection of cultured primary ovarian granulosa cells with 80 different constructs encoding human pre-miRNAs on the expression of the proliferation marker, PCNA, and the apoptosis marker, Bax was evaluated by immunocytochemistry. Eleven out of 80 tested miRNA constructs resulted in stimulation, and 53 miRNAs inhibited expression of PCNA. Furthermore, 11 of the 80 miRNAs tested promoted accumulation of Bax, while 46 miRNAs caused a reduction in Bax in human ovarian cells. In addition, two selected antisense constructs that block the corresponding miRNAs mir-15a and mir-188 were evaluated for their effects on expression of PCNA. An antisense construct inhibiting mir-15a (which precursor suppressed PCNA) increased PCNA, whereas an antisense construct for mir-188 (which precursor did not change PCNA) did not affect PCNA expression. Verification of effects of selected pre-mir-10a, mir-105, and mir-182 by using other markers of proliferation (cyclin B1) and apoptosis (TdT and caspase 3) confirmed specificity of miRNAs effects on these processes. This is the first direct demonstration of the involvement of miRNAs in controlling both proliferation and apoptosis by ovarian granulose cells, as well as the identification of miRNAs promoting and suppressing these processes utilizing a genome-wide miRNA screen.


Assuntos
Apoptose , Proliferação de Células , Células da Granulosa/metabolismo , MicroRNAs/metabolismo , Adulto , Caspase 3/metabolismo , Células Cultivadas , Ciclina B1/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Antígeno Nuclear de Célula em Proliferação/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Proteína X Associada a bcl-2/metabolismo
4.
J Cell Physiol ; 219(2): 415-20, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19194990

RESUMO

The aim of our studies was to identify miRNAs affecting the release of the major ovarian steroid hormones progestagen, androgen and estrogen by human ovarian cells. The effect of transfection of cultured primary ovarian granulosa cells with 80 different gene constructs encoding human pre-miRNAs on release of progesterone, testosterone and estradiol was evaluated by enzyme immunoassay. In addition, effect of two selected antisense constructs blocking corresponding miRNA on progesterone release was tested. Efficiency of transfection (incorporation transfection reagent) and silencing of marker substances (GAPDH mRNA, GAPDH and CREB-1) were validated by fluorescent microscopy, real-time reverse transcription-PCR analysis and immunocytochemical analysis. Thirty-six out of 80 tested miRNA constructs resulted in inhibition of progesterone release in granulosa cells, and 10 miRNAs promoted progesterone release. Transfected of cells with antisense constructs to two selected miRNAs blocking progesterone release induced increase in progesterone output. Fifty-seven miRNAs tested inhibited testosterone release, and only one miRNA enhanced testosterone output. Fifty-one miRNAs suppressed estradiol release, while none of the miRNAs tested stimulated it. This is the first demonstration that miRNAs can control reproductive functions resulting in enhanced or inhibited release of ovarian progestagen, androgen and estrogen. We hypothesize that such miRNA-mediated effects could be potentially used for regulation of reproductive processes, including fertility, and for treatment of reproductive and other steroid-dependent disorders.


Assuntos
Estrogênios , MicroRNAs , Ovário , Progestinas , Testosterona , Animais , Células Cultivadas , Estrogênios/genética , Estrogênios/metabolismo , Feminino , Inativação Gênica , Células da Granulosa/citologia , Células da Granulosa/fisiologia , Humanos , Imunoensaio , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Ovário/citologia , Ovário/metabolismo , Progestinas/genética , Progestinas/metabolismo , Precursores de RNA/genética , Precursores de RNA/metabolismo , Testosterona/genética , Testosterona/metabolismo , Transfecção
5.
Funct Integr Genomics ; 9(2): 185-95, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19067003

RESUMO

The aim of this study was to identify protein kinases (PKs) involved in the expression of proliferating cell nuclear antigen (PCNA) and p53, markers of proliferation and apoptosis in human ovarian cells. Cultured ovarian granulosa cells were subjected to transfection with 264 small-interfering RNA (siRNA) constructs from a siRNA library, which selectively blocked the expression of 88 known PKs. The efficiency of transfection and siRNA knockdown were validated by fluorescent microscopy, real-time reverse transcription polymerase chain reaction, and immunocytochemical analysis. The expression of PCNA and p53, before and after transfection with siRNA constructs, was evaluated by immunocytochemistry. The siRNA constructs suppressed the expression of their targets molecules by up to 84%. Knockdown of 32 of the 88 PKs inhibited the expression of PCNA, while the knockdown of seven of the PKs stimulated PCNA expression. Knockdown of 30 of the 88 PKs reduced the expression of p53, while knockdown of five PKs enhanced p53 expression. Our results illustrate that siRNA constructs are useful tools for understanding the role of PKs in the control of ovarian cell functions, such as proliferation and apoptosis. The specific knockdown of individual PKs has enabled the identification of a number of new PKs that control the expression of PCNA and p53 in human ovarian cells.


Assuntos
Células da Granulosa/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Adulto , Apoptose , Proliferação de Células , Células Cultivadas , Feminino , Células da Granulosa/citologia , Humanos , Técnicas In Vitro , Proteínas Quinases/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Transfecção
6.
Cancer Res ; 67(16): 7713-22, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17699775

RESUMO

MicroRNAs play important roles in animal development, cell differentiation, and metabolism and have been implicated in human cancer. The let-7 microRNA controls the timing of cell cycle exit and terminal differentiation in Caenorhabditis elegans and is poorly expressed or deleted in human lung tumors. Here, we show that let-7 is highly expressed in normal lung tissue, and that inhibiting let-7 function leads to increased cell division in A549 lung cancer cells. Overexpression of let-7 in cancer cell lines alters cell cycle progression and reduces cell division, providing evidence that let-7 functions as a tumor suppressor in lung cells. let-7 was previously shown to regulate the expression of the RAS lung cancer oncogenes, and our work now shows that multiple genes involved in cell cycle and cell division functions are also directly or indirectly repressed by let-7. This work reveals the let-7 microRNA to be a master regulator of cell proliferation pathways.


Assuntos
Neoplasias Hepáticas/genética , MicroRNAs/genética , Ciclo Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Quinase 6 Dependente de Ciclina/biossíntese , Quinase 6 Dependente de Ciclina/genética , Regulação Neoplásica da Expressão Gênica , Genes ras , Células HeLa , Humanos , Neoplasias Hepáticas/patologia , Pulmão/metabolismo , Pulmão/fisiologia , MicroRNAs/biossíntese , Análise em Microsséries , Transfecção , Fosfatases cdc25/biossíntese , Fosfatases cdc25/genética
7.
Cancer Biol Ther ; 20(9): 1281-1289, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31234707

RESUMO

Dichloroacetate (DCA) is an inhibitor of pyruvate dehydrogenase kinase (PDK) that has been shown to reverse the Warburg effect and cause tumor cell death. Clinical research into the anti-cancer activity of DCA revealed high dosage requirements and reports of toxicity. While there have been subsequent mechanistic investigations, a search for DCA alternatives could result in a safer and more effective anticancer therapy. This study evaluates eight small compounds with a conserved dichloric terminal and their in vitro and in vivo potential for anticancer activity. Initial viability screening across six cancer cell lines reveals even at 10 mg/mL, compound treatments do not result in complete cell death which suggests minimal compound cytotoxicity. Furthermore, in vivo data demonstrates that cationic dichloric compounds DCAH and DCMAH, which were selected for further testing based on highest in vitro viability impact, inhibit tumor growth in the U87 model of glioblastoma, suggesting their clinical potential as accessible anti-cancer drugs. Immunoblotting signaling data from tumor lysates demonstrates that the mechanism of actions of cationic DCAH and DCMAH are unlikely to be consistent with that of the terminally carboxylic DCA and warrants further independent investigation.


Assuntos
Antineoplásicos/farmacologia , Ácido Dicloroacético/farmacologia , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ácido Dicloroacético/química , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Biochem J ; 391(Pt 2): 441-8, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16060857

RESUMO

NGAL (human neutrophil gelatinase-associated lipocalin) and its mouse analogue 24p3 are members of the lipocalin family of small secreted proteins. These proteins are up-regulated in a number of pathological conditions, including cancers, and may function as transporters of essential factors. Although previous publications have suggested that 24p3 has pro-apoptotic functions, other data are more suggestive of a survival function. The current study was designed to determine whether NGAL is pro- or anti-apoptotic. Apoptosis induced in human adenocarcinoma A549 cells by the 5-lipoxygenase-activating-protein inhibitor MK886, or several celecoxib-derived PDK1 (phosphoinositide-dependent kinase 1) inhibitors that are devoid of cyclo-oxygenase-2 inhibitory activity, was accompanied by a dose- and time-dependent increase of NGAL mRNA levels, as was reported previously with 24p3. A similar induction of NGAL mRNA was observed in human breast cancer MCF7 cells treated with MK886, indicating this was not a cell-specific effect. Treatment of A549 cells with up to 150 mug/10(6) cells of purified recombinant NGAL protein had no effect on viability, whereas antisera against the full-length NGAL protein induced apoptosis in these cells. The stable overexpression of NGAL in A549 cells had no effect on proliferation or viability. However, the cell death induced by a PDK1 inhibitor was reduced by 50% in NGAL-overexpressing cells. Decreasing NGAL mRNA and protein expression with siRNA (small interfering RNA) in A549 cells increased the toxicity of a PDK1 inhibitor by approx. 45%. These data indicate that, although the induction of NGAL correlates with apoptosis, this induction represents a survival response. Because NGAL is a secreted protein, it may play an extracellular role in cell defence against toxicants and/or facilitate the survival of the remaining cells.


Assuntos
Proteínas de Fase Aguda/metabolismo , Apoptose , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Proteínas de Fase Aguda/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Indóis/farmacologia , Lipocalina-2 , Lipocalinas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Pirazóis/farmacologia , Pirazóis/toxicidade , RNA Mensageiro/metabolismo , Sulfonamidas/farmacologia , Sulfonamidas/toxicidade , Regulação para Cima/efeitos dos fármacos
9.
Methods Enzymol ; 392: 242-77, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15644186

RESUMO

During the past five years, RNA interference (RNAi) has emerged as arguably the best functional genomics tool available to date, providing direct, causal links between individual genes and loss-of-function phenotypes through robust, broadly applicable, and readily upscalable methodologies. Originally applied experimentally in C. elegans and Drosophila, RNAi is now widely used in mammalian cell systems also. The development of commercially available libraries of short interfering RNAs (siRNAs) and other RNAi silencing reagents targeting entire classes of human genes provide the opportunity to carry out genome-scale screens to discover and characterize gene functions directly in human cells. A key challenge of these studies, also faced by earlier genomics or proteomics approaches, resides in reaching an optimal balance between the necessarily high throughput and the desire to achieve the same level of detailed analysis that is routine in conventional small-scale studies. This chapter discusses technical aspects of how to perform such screens, what parameters to monitor, and which readouts to apply. Examples of homogenous assays and multiplexed high-content microscopy-based screens are demonstrated.


Assuntos
Genômica , Interferência de RNA , Apoptose , Automação , Divisão Celular , Linhagem Celular Tumoral , Genes Reporter , Humanos , RNA Interferente Pequeno/genética
10.
Microrna ; 3(1): 29-36, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25069510

RESUMO

Our study aimed to examine the role of micro RNA Mir15a in control of basic ovarian cell functions: proliferation, apoptosis, and secretory activity. In the first series of experiments, primary human ovarian granulosa cells were transfected with antisense construct blocking Mir15a (anti-Mir15a) and cultured without hormonal treatments. Accumulation of markers of proliferation (MAPK/ERK1,2 and PCNA) and apoptosis (caspase 3 and bax), and release of steroid hormones (progesterone, testosterone, and estradiol) were evaluated by immunocytochemical analysis and by enzyme immunoassay. In the second series of experiments, granulosa cells were transfected with gene construct encoding Mir15a precursor (pre-Mir15a) and cultured with and without follicle-stimulating hormone (FSH; 0, 1, 10, and 100 ng/ml). Expression of markers of proliferation (MAPK/ERK1,2) apoptosis (caspase 3), and steroidogenesis (release of progesterone, testosterone, and estradiol) were evaluated. Transfection of cells with anti-Mir15a resulted in a significant increase in accumulation of both proliferation and apoptosis markers, a reduction in progesterone and testosterone release, and an increase in estradiol release. Transfection of cells with pre-Mir15a had an opposite effect: it reduced accumulation of proliferation- and apoptosis-related proteins MAPK/ERK1,2 and caspase 3, and promoted release of progesterone and testosterone, but not estradiol. Moreover, pre-Mir15a reversed the effect of FSH on caspase 3, progesterone, and testosterone, but not on MAPK/ERK1,2 and estradiol. Our observations demonstrate involvement of Mir15a in control of multiple ovarian functions: proliferation, apoptosis, release of progesterone, androgen, and estrogen, and response to gonadotropin. Moreover, this is the first demonstration that miRNAs can affect response of cells to hormonal regulators. We propose that Mir15 could potentially be used for control of different reproductive processes.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/fisiologia , Hormônios/metabolismo , MicroRNAs/genética , Adulto , Apoptose , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , MicroRNAs/antagonistas & inibidores
11.
Int J Pharm ; 427(1): 134-42, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21856394

RESUMO

The development of short-interfering RNA (siRNA) offers new strategies for manipulating specific genes responsible for pathological disorders. Myriad cationic polymer and lipid formulations have been explored, but an effective, non-toxic carrier remains a major barrier to clinical translation. Among the emerging candidates for siRNA carriers are cell penetrating peptides (CPPs), which can traverse the plasma membrane and facilitate the intracellular delivery of siRNA. Previously, a highly efficient and non-cytotoxic means of gene delivery was designed by complexing plasmid DNA with CPPs, then condensing with calcium. Here, the CPP TAT and a longer, 'double' TAT (dTAT) were investigated as potential carriers for siRNA. Various N/P ratios and calcium concentrations were used to optimize siRNA complexes in vitro. Upon addition of calcium, 'loose' siRNA/CPP complexes were condensed into small nanoparticles. Knockdown of luciferase expression in the human epithelial lung cell line A549-luc-C8 was high (up to 93%) with no evidence of cytotoxicity. Selected formulations of the dTAT complexes were dosed intravenously up to 1000 mg/kg with minimal toxicity. Biodistribution studies revealed high levels of gene knockdown in the lung and muscle tissue suggesting these simple vectors may offer a translatable approach to siRNA delivery.


Assuntos
Peptídeos Penetradores de Células/química , Portadores de Fármacos/química , Inativação Gênica , Terapia Genética/métodos , RNA Interferente Pequeno/administração & dosagem , Cálcio/química , Linhagem Celular , Peptídeos Penetradores de Células/administração & dosagem , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/efeitos adversos , Humanos , Luciferases , Nanopartículas/administração & dosagem , Nanopartículas/química , RNA Interferente Pequeno/química , Distribuição Tecidual
12.
Exp Hematol ; 39(10): 1030-1042.e7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21784052

RESUMO

OBJECTIVE: The study investigated differential microRNA (miRNA) expression patterns in acute myeloid leukemia (AML) patients with intermediate-risk (IR) characteristics. After characterization and validation of miR-10a, which was specifically upregulated in nucleophosmin 1 (NPM1) mutant AML samples, functional consequences of miR-10a overexpression were further delineated in vitro. MATERIALS AND METHODS: Microarray analysis of miRNAs in bone marrow samples from AML (IR) patients with NPM1 mutations and healthy donors was performed to detect differential expression patterns. After validation of miRNA expression specific for NPM1 mutation in AML patients by quantitative reverse transcription polymerase chain reaction, a functional target gene search was conducted using complementary DNA microarray data from samples transfected with miR-10a. Potential target gene validation was done using transient transfection of K562 cells followed by Western blotting and luciferase reporter assay. RESULTS: In comparison with wild-type samples, NPM1 mutant AML samples were shown to markedly overexpress miR-10a. Subsequent in vitro miR-10a overexpression induced differential gene expression as determined by microarray analysis. Here the murine double minute 4 (MDM4) gene turned out as a candidate gene for miR-10a. Validation of MDM4 in leukemic cells revealed a robust negative relationship between miR-10a overexpression and MDM4 downregulation. Furthermore, we determined an inverse association between miR-10a and MDM4 expression in AML (IR) samples with respect to their NPM1 mutational status. CONCLUSIONS: miR-10a expression is highly characteristic for AML (IR) patients with NPM1 mutations and may influence its biological properties in AML by interfering with the p53 machinery partly regulated by MDM4.


Assuntos
Regulação Leucêmica da Expressão Gênica/genética , Leucemia Mieloide/metabolismo , MicroRNAs/fisiologia , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , RNA Neoplásico/fisiologia , Regiões 3' não Traduzidas , Doença Aguda , Idoso , Medula Óssea/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/patologia , Masculino , MicroRNAs/biossíntese , MicroRNAs/genética , Pessoa de Meia-Idade , Mutação , Proteínas de Neoplasias/biossíntese , Proteínas Nucleares/biossíntese , Nucleofosmina , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas/biossíntese , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Risco , Transfecção , Células Tumorais Cultivadas/metabolismo , Proteína Supressora de Tumor p53/fisiologia
13.
J Mol Endocrinol ; 44(1): 45-53, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19713423

RESUMO

The goal of this study was to identify protein kinases (PKs) that control the secretory activity of human ovarian cells. Cultured ovarian granulosa cells were transfected with 264 siRNA constructs that selectively block the expression of 88 known PKs. The efficiency of transfection and of silencing marker molecules (glyceraldehyde 3-phosphate dehydrogenase, GAPDH and CDC2/p34 PK) was validated by fluorescence microscopy, real-time reverse transcription-PCR, and immunocytochemistry. Release of steroid hormones (progesterone, P(4)) and IGF1 was determined by RIA. siRNA suppressed the expression of marker molecules by up to 84%. P(4) release was suppressed after inhibiting 34 individual PKs and was stimulated after inhibiting 12 PKs. Blocking nine individual PKs inhibited IGF1 release, while the inactivation of 17 others stimulated IGF1 release. Together, these results demonstrate that the release of both steroid and peptide hormones by human ovarian cells is controlled by a large number of PKs, and that siRNA constructs may be useful tools for further defining the role of PKs in controlling ovarian secretory function.


Assuntos
Hormônios Gonadais/metabolismo , Ovário/metabolismo , Proteínas Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Animais , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Células Cultivadas , Feminino , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Proteínas Quinases/genética , RNA Interferente Pequeno/genética
14.
Cancer Res ; 67(22): 10782-8, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18006822

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) binds to death receptors 4/5 and selectively induces caspase-dependent apoptosis. The RNA interference screening approach has led to the discovery and characterization of several TRAIL pathway components in human cells. Here, libraries of synthetic small interfering RNA (siRNA) and microRNAs (miRNA) were used to probe the TRAIL pathway. In addition to known genes, siRNAs targeting CDK4, PTGS1, ALG2, CLCN3, IRAK4, and MAP3K8 altered TRAIL-induced caspase-3 activation responses. Introduction of the miRNAs let-7c, mir-10a, mir-144, mir-150, mir-155, and mir-193 also affected the activation of the caspase cascade. Putative targets of these endogenous miRNAs included genes encoding death receptors, caspases, and other apoptosis-related genes. Among the novel genes revealed in the screen, CDK4 was selected for further characterization. CDK4 was the only member of the cyclin-dependent kinase gene family that bore a unique function in apoptotic signal transduction.


Assuntos
Apoptose , Quinase 4 Dependente de Ciclina/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , RNA/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Inativação Gênica , Genoma Humano , Humanos , MicroRNAs/metabolismo , Modelos Biológicos , RNA Interferente Pequeno/metabolismo
15.
RNA ; 11(6): 985-93, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15923380

RESUMO

Small interfering RNAs (siRNAs) are being used to induce sequence-specific gene silencing in cultured cells to study mammalian gene function. Libraries of siRNAs targeting entire human gene classes can be used to identify genes with specific cellular functions. Here we describe high-throughput siRNA delivery methods to facilitate siRNA library screening experiments with both immortalized and primary cells. We adapted chemical reverse transfection for immortalized adherent cell lines in a 96-well format. The method is fast, robust, and exceptionally effective for many cell types. For primary cells and immortalized cells that are recalcitrant to lipofection-based methods, we developed electropermeabilization (electroporation) conditions that facilitate siRNA delivery to a broad range of cell types, including primary human T-cells, hMSC, NHA, NDHF-Neo, HUVEC, DI TNC1, RPTEC, PC12, and K562 cells. To enable high-throughput electropermeabilization of primary cells, we developed a novel 96-well electroporation device that provides highly efficient and reproducible delivery of siRNAs. The combination of high-throughput chemical reverse transfection and electroporation makes it possible to deliver libraries of siRNAs to virtually any cell type, enabling gene function analysis and discovery on a genome scale.


Assuntos
Eletroporação/métodos , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Transfecção/métodos , Animais , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Biblioteca Gênica , Humanos , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/farmacologia , RNA Interferente Pequeno/genética , Ratos
16.
Genome Res ; 15(7): 928-35, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15965026

RESUMO

Mutations in distant regulatory elements can have a negative impact on human development and health, yet because of the difficulty of detecting these critical sequences, we predominantly focus on coding sequences for diagnostic purposes. We have undertaken a comparative sequence-based approach to characterize a large noncoding region deleted in patients affected by Van Buchem (VB) disease, a severe sclerosing bone dysplasia. Using BAC recombination and transgenesis, we characterized the expression of human sclerostin (SOST) from normal (SOST(wt)) or Van Buchem (SOST(vbDelta) alleles. Only the SOST(wt) allele faithfully expressed high levels of human SOST in the adult bone and had an impact on bone metabolism, consistent with the model that the VB noncoding deletion removes a SOST-specific regulatory element. By exploiting cross-species sequence comparisons with in vitro and in vivo enhancer assays, we were able to identify a candidate enhancer element that drives human SOST expression in osteoblast-like cell lines in vitro and in the skeletal anlage of the embryonic day 14.5 (E14.5) mouse embryo, and discovered a novel function for sclerostin during limb development. Our approach represents a framework for characterizing distant regulatory elements associated with abnormal human phenotypes.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Elementos Facilitadores Genéticos , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Marcadores Genéticos/genética , Hiperostose/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Densidade Óssea/genética , Ligação Genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Ratos , Alinhamento de Sequência , Células Tumorais Cultivadas , Regiões não Traduzidas
17.
Science ; 299(5611): 1391-4, 2003 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-12610304

RESUMO

Nonhuman primates represent the most relevant model organisms to understand the biology of Homo sapiens. The recent divergence and associated overall sequence conservation between individual members of this taxon have nonetheless largely precluded the use of primates in comparative sequence studies. We used sequence comparisons of an extensive set of Old World and New World monkeys and hominoids to identify functional regions in the human genome. Analysis of these data enabled the discovery of primate-specific gene regulatory elements and the demarcation of the exons of multiple genes. Much of the information content of the comprehensive primate sequence comparisons could be captured with a small subset of phylogenetically close primates. These results demonstrate the utility of intraprimate sequence comparisons to discover common mammalian as well as primate-specific functional elements in the human genome, which are unattainable through the evaluation of more evolutionarily distant species.


Assuntos
Genoma Humano , Genoma , Filogenia , Primatas/genética , Análise de Sequência de DNA , Animais , Apolipoproteínas A/genética , Evolução Biológica , Cebidae/genética , Cercopithecidae/genética , Biologia Computacional , Sequência Conservada , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Éxons , Regulação da Expressão Gênica , Hominidae/genética , Humanos , Hylobates/genética , Funções Verossimilhança , Sequências Reguladoras de Ácido Nucleico , Especificidade da Espécie , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA