Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Genes Dev ; 25(16): 1734-45, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21816922

RESUMEN

The miR-17~92 cluster is a potent microRNA-encoding oncogene. Here, we show that miR-17~92 synergizes with loss of Rb family members to promote retinoblastoma. We observed miR-17~92 genomic amplifications in murine retinoblastoma and high expression of miR-17~92 in human retinoblastoma. While miR-17~92 was dispensable for mouse retinal development, miR-17~92 overexpression, together with deletion of Rb and p107, led to rapid emergence of retinoblastoma with frequent metastasis to the brain. miR-17~92 oncogenic function in retinoblastoma was not mediated by a miR-19/PTEN axis toward apoptosis suppression, as found in lymphoma/leukemia models. Instead, miR-17~92 increased the proliferative capacity of Rb/p107-deficient retinal cells. We found that deletion of Rb family members led to compensatory up-regulation of the cyclin-dependent kinase inhibitor p21Cip1. miR-17~92 overexpression counteracted p21Cip1 up-regulation, promoted proliferation, and drove retinoblastoma formation. These results demonstrate that the oncogenic determinants of miR-17~92 are context-specific and provide new insights into miR-17~92 function as an RB-collaborating gene in cancer.


Asunto(s)
MicroARNs/genética , Mutación , Proteína de Retinoblastoma/genética , Retinoblastoma/genética , Transducción de Señal/genética , Animales , Animales Recién Nacidos , Línea Celular Tumoral , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Familia de Multigenes , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos Antisentido/genética , Embarazo , Retina/embriología , Retina/crecimiento & desarrollo , Retina/metabolismo , Retinoblastoma/metabolismo , Retinoblastoma/patología , Proteína de Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/metabolismo
2.
Nat Genet ; 38(2): 228-33, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16380711

RESUMEN

Understanding the molecular mechanisms that regulate cellular proliferation and differentiation is a central theme of developmental biology. MicroRNAs (miRNAs) are a class of regulatory RNAs of approximately 22 nucleotides that post-transcriptionally regulate gene expression. Increasing evidence points to the potential role of miRNAs in various biological processes. Here we show that miRNA-1 (miR-1) and miRNA-133 (miR-133), which are clustered on the same chromosomal loci, are transcribed together in a tissue-specific manner during development. miR-1 and miR-133 have distinct roles in modulating skeletal muscle proliferation and differentiation in cultured myoblasts in vitro and in Xenopus laevis embryos in vivo. miR-1 promotes myogenesis by targeting histone deacetylase 4 (HDAC4), a transcriptional repressor of muscle gene expression. By contrast, miR-133 enhances myoblast proliferation by repressing serum response factor (SRF). Our results show that two mature miRNAs, derived from the same miRNA polycistron and transcribed together, can carry out distinct biological functions. Together, our studies suggest a molecular mechanism in which miRNAs participate in transcriptional circuits that control skeletal muscle gene expression and embryonic development.


Asunto(s)
Diferenciación Celular , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Embrión no Mamífero/citología , Expresión Génica , Ratones , Modelos Biológicos , Mioblastos/citología , Mioblastos/metabolismo , Miocardio/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Xenopus/embriología
3.
Nat Genet ; 37(6): 630-5, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15864308

RESUMEN

Modern yeast living in fleshy fruits rapidly convert sugars into bulk ethanol through pyruvate. Pyruvate loses carbon dioxide to produce acetaldehyde, which is reduced by alcohol dehydrogenase 1 (Adh1) to ethanol, which accumulates. Yeast later consumes the accumulated ethanol, exploiting Adh2, an Adh1 homolog differing by 24 (of 348) amino acids. As many microorganisms cannot grow in ethanol, accumulated ethanol may help yeast defend resources in the fruit. We report here the resurrection of the last common ancestor of Adh1 and Adh2, called Adh(A). The kinetic behavior of Adh(A) suggests that the ancestor was optimized to make (not consume) ethanol. This is consistent with the hypothesis that before the Adh1-Adh2 duplication, yeast did not accumulate ethanol for later consumption but rather used Adh(A) to recycle NADH generated in the glycolytic pathway. Silent nucleotide dating suggests that the Adh1-Adh2 duplication occurred near the time of duplication of several other proteins involved in the accumulation of ethanol, possibly in the Cretaceous age when fleshy fruits arose. These results help to connect the chemical behavior of these enzymes through systems analysis to a time of global ecosystem change, a small but useful step towards a planetary systems biology.


Asunto(s)
Alcohol Deshidrogenasa/metabolismo , Evolución Biológica , Saccharomyces cerevisiae/metabolismo , Alcohol Deshidrogenasa/genética , Secuencia de Bases , Etanol/metabolismo , Datos de Secuencia Molecular
4.
Biochem Biophys Res Commun ; 440(1): 99-104, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-24045012

RESUMEN

Recent progress has been made in the identification of protein-coding genes and miRNAs that are expressed in and alter the behavior of colonic epithelia. However, the role of long non-coding RNAs (lncRNAs) in colonic homeostasis is just beginning to be explored. By gene expression profiling of post-mitotic, differentiated tops and proliferative, progenitor-compartment bottoms of microdissected adult mouse colonic crypts, we identified several lncRNAs more highly expressed in crypt bottoms. One identified lncRNA, designated non-coding Nras functional RNA (ncNRFR), resides within the Nras locus but appears to be independent of the Nras coding transcript. Stable overexpression of ncNRFR in non-transformed, conditionally immortalized mouse colonocytes results in malignant transformation, as determined by growth in soft agar and formation of highly invasive tumors in nude mice. Moreover, ncNRFR appears to inhibit the function of the tumor suppressor let-7. These results suggest precise regulation of ncNRFR is necessary for proper cell growth in the colonic crypt, and its misregulation results in neoplastic transformation.


Asunto(s)
Transformación Celular Neoplásica , Colon/patología , Neoplasias del Colon/genética , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Animales , Colon/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Ratones , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo
5.
Nature ; 435(7043): 828-33, 2005 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-15944707

RESUMEN

To date, more than 200 microRNAs have been described in humans; however, the precise functions of these regulatory, non-coding RNAs remains largely obscure. One cluster of microRNAs, the mir-17-92 polycistron, is located in a region of DNA that is amplified in human B-cell lymphomas. Here we compared B-cell lymphoma samples and cell lines to normal tissues, and found that the levels of the primary or mature microRNAs derived from the mir-17-92 locus are often substantially increased in these cancers. Enforced expression of the mir-17-92 cluster acted with c-myc expression to accelerate tumour development in a mouse B-cell lymphoma model. Tumours derived from haematopoietic stem cells expressing a subset of the mir-17-92 cluster and c-myc could be distinguished by an absence of apoptosis that was otherwise prevalent in c-myc-induced lymphomas. Together, these studies indicate that non-coding RNAs, specifically microRNAs, can modulate tumour formation, and implicate the mir-17-92 cluster as a potential human oncogene.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes/genética , Linfoma de Células B/genética , MicroARNs/genética , Oncogenes/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Genes myc/genética , Humanos , Ratones , Datos de Secuencia Molecular , Familia de Multigenes/genética , Fenotipo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
6.
RNA ; 14(8): 1539-49, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18566191

RESUMEN

A hallmark of mammalian embryonic development is the widespread induction of microRNA (miRNA) expression. Surprisingly, the transcription of many of these small, noncoding RNAs is unchanged through development; rather, a post-transcriptional regulatory event prevents accumulation of the mature miRNA species. Here, we present a biochemical framework for the regulated production of the Let-7 family of miRNAs. Embryonic cells contain a Drosha Inhibitor that prevents processing of the Let-7 primary transcript. This inhibitor specifically binds to conserved nucleotides in the loop region of the Let-7 precursor, and competitor RNAs that mimic the binding site restore Let-7 processing. We have identified the Drosha Inhibitor as the embryonic stem cell specific protein Lin-28. Lin-28 has been previously implicated in developmental regulatory pathways in Caenorhabditis elegans, and it promotes reprogramming of human somatic cells into pluripotent stem cells. Our findings outline a microRNA post-transcriptional regulatory network and establish a novel role for the miRNA precursor loop in the regulated production of mature Let-7.


Asunto(s)
MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular , Sistema Libre de Células , Células HeLa , Humanos , Ratones , Interferencia de ARN
7.
Nature ; 425(6955): 285-8, 2003 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-13679914

RESUMEN

Features of the physical environment surrounding an ancestral organism can be inferred by reconstructing sequences of ancient proteins made by those organisms, resurrecting these proteins in the laboratory, and measuring their properties. Here, we resurrect candidate sequences for elongation factors of the Tu family (EF-Tu) found at ancient nodes in the bacterial evolutionary tree, and measure their activities as a function of temperature. The ancient EF-Tu proteins have temperature optima of 55-65 degrees C. This value seems to be robust with respect to uncertainties in the ancestral reconstruction. This suggests that the ancient bacteria that hosted these particular genes were thermophiles, and neither hyperthermophiles nor mesophiles. This conclusion can be compared and contrasted with inferences drawn from an analysis of the lengths of branches in trees joining proteins from contemporary bacteria, the distribution of thermophily in derived bacterial lineages, the inferred G + C content of ancient ribosomal RNA, and the geological record combined with assumptions concerning molecular clocks. The study illustrates the use of experimental palaeobiochemistry and assumptions about deep phylogenetic relationships between bacteria to explore the character of ancient life.


Asunto(s)
Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Ambiente , Factor Tu de Elongación Peptídica/metabolismo , Bacterias/clasificación , Composición de Base , Estabilidad de Enzimas , Guanosina Difosfato/metabolismo , Datos de Secuencia Molecular , Filogenia , Unión Proteica , ARN Ribosómico/genética , Temperatura , Factores de Tiempo
8.
Mol Biol Cell ; 16(10): 4941-53, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16079180

RESUMEN

When Pichia pastoris adapts from methanol to glucose growth, peroxisomes are rapidly sequestered and degraded within the vacuole by micropexophagy. During micropexophagy, sequestering membranes arise from the vacuole to engulf the peroxisomes. Fusion of the sequestering membranes and incorporation of the peroxisomes into the vacuole is mediated by the micropexophagy-specific membrane apparatus (MIPA). In this study, we show the P. pastoris ortholog of Atg9, a novel membrane protein is essential for the formation of the sequestering membranes and assembly of MIPA. During methanol growth, GFP-PpAtg9 localizes to multiple structures situated near the plasma membrane referred as the peripheral compartment (Atg9-PC). On glucose-induced micropexophagy, PpAtg9 traffics from the Atg9-PC to unique perivacuolar structures (PVS) that contain PpAtg11, but lack PpAtg2 and PpAtg8. Afterward, PpAtg9 distributes to the vacuole surface and sequestering membranes. Movement of the PpAtg9 from the Atg9-PC to the PVS requires PpAtg11 and PpVps15. PpAtg2 and PpAtg7 are essential for PpAtg9 trafficking from the PVS to the vacuole and sequestering membranes, whereas trafficking of PpAtg9 proceeds independent of PpAtg1, PpAtg18, and PpVac8. In summary, our data suggest that PpAtg9 transits from the Atg9-PC to the PVS and then to the sequestering membranes that engulf the peroxisomes for degradation.


Asunto(s)
Proteínas Fúngicas/metabolismo , Membranas Intracelulares/fisiología , Proteínas de la Membrana/metabolismo , Peroxisomas/fisiología , Pichia/fisiología , Vacuolas/fisiología , Proteínas Fúngicas/genética , Glucosa/metabolismo , Membranas Intracelulares/ultraestructura , Metanol/metabolismo , Microscopía Electrónica de Transmisión , Peroxisomas/ultraestructura , Pichia/metabolismo , Pichia/ultraestructura , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Vacuolas/ultraestructura
9.
Methods Enzymol ; 427: 107-22, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17720481

RESUMEN

MicroRNAs (miRNAs) are small, noncoding RNAs that regulate the expression of target mRNAs. Although thousands of miRNAs have been identified, few have been functionally linked to specific biological pathways. Microarray-based expression analysis is an ideal strategy for identifying candidate miRNAs that correlate with biological pathways and for generating molecular signatures of disease states. This chapter will describe a simple, low-cost microarray platform optimized for miRNA expression analysis.


Asunto(s)
Perfilación de la Expresión Génica , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Hibridación de Ácido Nucleico
10.
Front Microbiol ; 5: 565, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25400626

RESUMEN

DNA polymerases have evolved for billions of years to accept natural nucleoside triphosphate substrates with high fidelity and to exclude closely related structures, such as the analogous ribonucleoside triphosphates. However, polymerases that can accept unnatural nucleoside triphosphates are desired for many applications in biotechnology. The focus of this review is on non-standard nucleotides that expand the genetic "alphabet." This review focuses on experiments that, by directed evolution, have created variants of DNA polymerases that are better able to accept unnatural nucleotides. In many cases, an analysis of past evolution of these polymerases (as inferred by examining multiple sequence alignments) can help explain some of the mutations delivered by directed evolution.

11.
PLoS One ; 8(1): e53436, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23335963

RESUMEN

Overexpression of ribonucleotide reductase subunit M2 (RRM2), involved in deoxyribonucleotide synthesis, drives the chemoresistance of pancreatic cancer to nucleoside analogs (e.g., gemcitabine). While silencing RRM2 by synthetic means has shown promise in reducing chemoresistance, targeting endogenous molecules, especially microRNAs (miRNAs), to advance chemotherapeutic outcomes has been poorly explored. Based on computational predictions, we hypothesized that the let-7 tumor suppressor miRNAs will inhibit RRM2-mediated gemcitabine chemoresistance in pancreatic cancer. Reduced expression of the majority of let-7 miRNAs with an inverse relationship to RRM2 expression was identified in innately gemcitabine-resistant pancreatic cancer cell lines. Direct binding of let-7 miRNAs to the 3' UTR of RRM2 transcripts identified post-transcriptional regulation of RRM2 influencing gemcitabine chemosensitivity. Intriguingly, overexpression of human precursor-let-7 miRNAs led to differential RRM2 expression and chemosensitivity responses in a poorly differentiated pancreatic cancer cell line, MIA PaCa-2. Defective processing of let-7a precursors to mature forms, in part, explained the discrepancies observed with let-7a expressional outcomes. Consistently, the ratios of mature to precursor let-7a were progressively reduced in gemcitabine-sensitive L3.6pl and Capan-1 cell lines induced to acquire gemcitabine resistance. Besides known regulators of let-7 biogenesis (e.g., LIN-28), short hairpin RNA library screening identified several novel RNA binding proteins, including the SET oncoprotein, to differentially impact let-7 biogenesis and chemosensitivity in gemcitabine-sensitive versus -resistant pancreatic cancer cells. Further, LIN-28 and SET knockdown in the cells led to profound reductions in cellular proliferation and colony-formation capacities. Finally, defective processing of let-7a precursors with a positive correlation to RRM2 overexpression was identified in patient-derived pancreatic ductal adenocarcinoma (PDAC) tissues. These data demonstrate an intricate post-transcriptional regulation of RRM2 and chemosensitivity by let-7a and that the manipulation of regulatory proteins involved in let-7a transcription/processing may provide a mechanism for improving chemotherapeutic and/or tumor growth control responses in pancreatic cancer.


Asunto(s)
Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias Pancreáticas/genética , Ribonucleósido Difosfato Reductasa/genética , Antineoplásicos/farmacología , Línea Celular , Células Cultivadas , Proteínas de Unión al ADN , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Expresión Génica , Técnicas de Silenciamiento del Gen , Chaperonas de Histonas/metabolismo , Humanos , MicroARNs/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Interferencia de ARN , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Gemcitabina
12.
PLoS One ; 5(1): e8742, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20090903

RESUMEN

BACKGROUND: The apicomplexan parasite Toxoplasma gondii can infect and replicate in virtually any nucleated cell in many species of warm-blooded animals; thus, it has evolved the ability to exploit well-conserved biological processes common to its diverse hosts. Here we have investigated whether Toxoplasma modulates the levels of host microRNAs (miRNAs) during infection. METHODOLOGY/PRINCIPAL FINDINGS: Using microarray profiling and a combination of conventional molecular approaches we report that Toxoplasma specifically modulates the expression of important host microRNAs during infection. We show that both the primary transcripts for miR-17 approximately 92 and miR-106b approximately 25 and the pivotal miRNAs that are derived from miR-17 approximately 92 display increased abundance in Toxoplasma-infected primary human cells; a Toxoplasma-dependent up-regulation of the miR-17 approximately 92 promoter is at least partly responsible for this increase. The abundance of mature miR-17 family members, which are derived from these two miRNA clusters, remains unchanged in host cells infected with the closely related apicomplexan Neospora caninum; thus, the Toxoplasma-induced increase in their abundance is a highly directed process rather than a general host response to infection. CONCLUSIONS/SIGNIFICANCE: Altered levels of miR-17 approximately 92 and miR-106b approximately 25 are known to play crucial roles in mammalian cell regulation and have been implicated in numerous hyperproliferative diseases although the mechanisms driving their altered expression are unknown. Hence, in addition to the implications of these findings on the host-pathogen interaction, Toxoplasma may represent a powerful probe for understanding the normal mechanisms that regulate the levels of key host miRNAs.


Asunto(s)
Interacciones Huésped-Patógeno , MicroARNs/metabolismo , Toxoplasma/patogenicidad , Toxoplasmosis/genética , Northern Blotting , Células Cultivadas , Humanos , Regiones Promotoras Genéticas , Toxoplasmosis/parasitología , Transcripción Genética
13.
Genes Dev ; 22(6): 728-33, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18347091

RESUMEN

Appendage regeneration is defined by rapid changes in gene expression that achieve dramatic developmental effects, suggesting involvement of microRNAs (miRNAs). Here, we find dynamic regulation of many miRNAs during zebrafish fin regeneration. In particular, miR-133 levels are high in uninjured fins but low during regeneration. When regeneration was blocked by Fibroblast growth factor (Fgf) receptor inhibition, high miR-133 levels were quickly restored. Experimentally increasing amounts of miR-133 attenuated fin regeneration. Conversely, miR-133 antagonism during Fgf receptor inhibition accelerated regeneration through increased proliferation within the regeneration blastema. The Mps1 kinase, an established positive regulator of blastemal proliferation, is an in vivo target of miR-133. Our findings identify miRNA depletion as a new regulatory mechanism for complex tissue regeneration.


Asunto(s)
Extremidades/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , MicroARNs/fisiología , Regeneración/fisiología , Pez Cebra/fisiología , Animales , Northern Blotting , Proliferación Celular , Embrión no Mamífero/citología , Embrión no Mamífero/fisiología , Técnica del Anticuerpo Fluorescente , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Pez Cebra/metabolismo
14.
J Biol Chem ; 282(4): 2130-4, 2007 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-17135268

RESUMEN

Micro-RNAs (miRNAs) are a class of non-coding RNAs that post-transcriptionally regulate gene expression via the RNA interference pathway. In addition to roles in normal development, miRNAs have recently been implicated in a range of human diseases, including cancer. We recently demonstrated that a polycistronic cluster of miRNAs, miR-17-92, is oncogenic in a mouse model for Burkitt's lymphoma. This is due, in part, to a reduced apoptotic program. In an effort to understand the regulation of miR-17-92, we have studied the promoter structure of this miRNA cluster. The primary transcript initiates from a consensus initiator sequence downstream of a nonconsensus TATA box. The core promoter region contains two functional E2F transcription factor binding sites. Chromatin immunoprecipitation demonstrates that E2F3 is the primary E2F family member that occupies the promoter. These data place miR-17-92 in a regulatory loop between E2F3 and the miR-17 target E2F1. We propose a model whereby miR-17-92 promotes cell proliferation by shifting the E2F transcriptional balance away from the pro-apoptotic E2F1 and toward the proliferative E2F3 transcriptional network.


Asunto(s)
Factores de Transcripción E2F/genética , Regulación de la Expresión Génica , MicroARNs/genética , Células 3T3 , Animales , Secuencia de Bases , Factores de Transcripción E2F/metabolismo , Humanos , Ratones , MicroARNs/metabolismo , Modelos Genéticos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas
15.
Dev Biol ; 310(2): 442-53, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17765889

RESUMEN

The miR-17-92 locus encodes a cluster of 7 microRNAs transcribed as a single primary transcript. It can accelerate c-Myc induced B cell lymphoma development and is highly expressed in many tumors, including lung tumors. However, the role of miR-17-92 in development has not been well studied. From analysis of microRNAs during lung development, expression of the miR-17-92 cluster is high at early stages, but declines as development proceeds. We used the mouse surfactant protein C (Sftpc) promoter to over-express the cluster in embryonic lung epithelium. Transgenic lungs have a very abnormal lethal phenotype. They contain numerous proliferative epithelial cells that retain high levels of Sox9, a marker of distal progenitors. The differentiation of proximal epithelial cells was also inhibited. Furthermore, a significant increase in the number of neuroendocrine cell clusters was observed in the lungs of dead transgenic pups. We identify a tumor suppressor, Rbl2 which belongs to the Rb family, as a new target for miR-17-5p. Together, these studies suggest that mir-17-92 normally promotes the high proliferation and undifferentiated phenotype of lung epithelial progenitor cells.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Epiteliales/citología , Pulmón/citología , MicroARNs/biosíntesis , Células Madre/citología , Animales , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Pulmón/embriología , Pulmón/metabolismo , Ratones , Ratones Transgénicos , Péptidos/genética , Péptidos/metabolismo , Regiones Promotoras Genéticas , Proteína C Asociada a Surfactante Pulmonar , Células Madre/metabolismo
16.
J Mol Cell Cardiol ; 42(6): 1137-41, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17498736

RESUMEN

MicroRNAs (miRNAs) are a recently discovered class of approximately 22-nucleotide regulatory RNAs that post-transcriptionally regulate gene expression. We have recently demonstrated that muscle-specific miRNAs miR-1 and miR-133 play an important role in modulating muscle proliferation and differentiation. Here, we investigate the involvement of miRNAs in cardiac hypertrophy. We analyzed the global expression of miRNAs in agonist-induced hypertrophic cardiomyocytes as well as in pressure overload-induced hypertrophic hearts and found the miRNA expression profile altered in those hypertrophic conditions. We further show that inhibition of endogenous miR-21 or miR-18b augments hypertrophic growth. Conversely, introduction of functional miR-21 or miR-18b into cardiomyocytes represses myocyte hypertrophy. Together, our studies point to miRNAs as critical regulators of cardiac hypertrophy.


Asunto(s)
Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Miocitos Cardíacos/patología , Animales , Animales Recién Nacidos , Células Cultivadas , Hipertrofia/metabolismo , Hipertrofia/patología , Hipertrofia/fisiopatología , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Ratas , Transfección
17.
Genome Biol ; 8(2): R27, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17326821

RESUMEN

BACKGROUND: microRNAs (miRNAs) are small, noncoding RNA molecules that are now thought to regulate the expression of many mRNAs. They have been implicated in the etiology of a variety of complex diseases, including Tourette's syndrome, Fragile x syndrome, and several types of cancer. RESULTS: We hypothesized that schizophrenia might be associated with altered miRNA profiles. To investigate this possibility we compared the expression of 264 human miRNAs from postmortem prefrontal cortex tissue of individuals with schizophrenia (n = 13) or schizoaffective disorder (n = 2) to tissue of 21 psychiatrically unaffected individuals using a custom miRNA microarray. Allowing a 5% false discovery rate, we found that 16 miRNAs were differentially expressed in prefrontal cortex of patient subjects, with 15 expressed at lower levels (fold change 0.63 to 0.89) and 1 at a higher level (fold change 1.77) than in the psychiatrically unaffected comparison subjects. The expression levels of 12 selected miRNAs were also determined by quantitative RT-PCR in our lab. For the eight miRNAs distinguished by being expressed at lower microarray levels in schizophrenia samples versus comparison samples, seven were also expressed at lower levels with quantitative RT-PCR. CONCLUSION: This study is the first to find altered miRNA profiles in postmortem prefrontal cortex from schizophrenia patients.


Asunto(s)
MicroARNs/metabolismo , Corteza Prefrontal/metabolismo , Trastornos Psicóticos/genética , Esquizofrenia/genética , Secuencias de Aminoácidos/genética , Cartilla de ADN , Femenino , Humanos , Masculino , MicroARNs/genética , Análisis por Micromatrices , Trastornos Psicóticos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esquizofrenia/metabolismo
18.
Autophagy ; 2(4): 280-8, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16921262

RESUMEN

We have identified the Pichia pastoris Vac8 homolog, a 60-64 kDa armadillo repeat protein, and have examined the role of PpVac8 in the degradative pathways involving the yeast vacuole. We report here that PpVac8 is required for glucose-induced pexophagy, but not ethanol-induced pexophagy or starvation-induced autophagy. This has been demonstrated by the persistence of peroxisomal alcohol oxidase activity in mutants lacking PpVac8 during glucose adaptation. During glucose-induced micropexophagy, in the absence of PpVac8, the vacuole was invaginated with arm-like "segmented" extensions that almost completely surrounded the adjacent peroxisomes. Vac8-GFP was found at the vacuolar membrane and concentrated at the base of the arm-like protrusions that extend from the vacuole to sequester the peroxisomes. The localization of Vac8-GFP to the vacuolar membrane occurred independent of PpAtg1, PpAtg9 or PpAtg11. Mutagenesis of the palmitoylated cysteines to alanines or deletion of the myristoylation and palmitoylation sites of PpVac8 resulted in decreased protein stability, impaired vacuolar association and reduced degradation of peroxisomal alcohol oxidase. Deletion of the central armadillo repeat domains of the PpVac8 did not alter its association with the vacuolar membrane, but resulted in a non-functional protein that suppressed the formation of the arm-like extensions from the vacuole to engulf the peroxisomes. PpVac8 is essential for the trafficking of PpAtg11, but not PpAtg1 or PpAtg18, to the vacuole membrane. Together, our results support a role for PpVac8 in early (formation of sequestering membranes) and late (post-MIPA membrane fusion) molecular events of glucose-induced pexophagy.


Asunto(s)
Autofagia/fisiología , Proteínas Fúngicas/metabolismo , Glucosa/metabolismo , Pichia/fisiología , Oxidorreductasas de Alcohol/metabolismo , Proteínas Fúngicas/genética , Pichia/citología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Vacuolas/metabolismo , Vacuolas/ultraestructura , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
19.
Genes Dev ; 20(16): 2202-7, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16882971

RESUMEN

MicroRNAs (miRNAs) are short, noncoding RNAs that post-transcriptionally regulate gene expression. While hundreds of mammalian miRNA genes have been identified, little is known about the pathways that regulate the production of active miRNA species. Here we show that a large fraction of miRNA genes are regulated post-transcriptionally. During early mouse development, many miRNA primary transcripts, including the Let-7 family, are present at high levels but are not processed by the enzyme Drosha. An analysis of gene expression in primary tumors indicates that the widespread down-regulation of miRNAs observed in cancer is due to a failure at the Drosha processing step. These data uncover a novel regulatory step in miRNA function and provide a mechanism for miRNA down-regulation in cancer.


Asunto(s)
Embrión de Mamíferos/metabolismo , MicroARNs/metabolismo , Neoplasias/genética , Procesamiento Postranscripcional del ARN , Animales , Northern Blotting , Línea Celular , Línea Celular Tumoral , Regulación hacia Abajo , Desarrollo Embrionario , Femenino , Ratones , MicroARNs/genética , Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasa III/metabolismo , Células Madre/metabolismo , Teratocarcinoma/genética , Teratocarcinoma/metabolismo , Regulación hacia Arriba
20.
Science ; 308(5723): 833-8, 2005 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-15774722

RESUMEN

MicroRNAs (miRNAs) are small RNAs that regulate gene expression posttranscriptionally. To block all miRNA formation in zebrafish, we generated maternal-zygotic dicer (MZdicer) mutants that disrupt the Dicer ribonuclease III and double-stranded RNA-binding domains. Mutant embryos do not process precursor miRNAs into mature miRNAs, but injection of preprocessed miRNAs restores gene silencing, indicating that the disrupted domains are dispensable for later steps in silencing. MZdicer mutants undergo axis formation and differentiate multiple cell types but display abnormal morphogenesis during gastrulation, brain formation, somitogenesis, and heart development. Injection of miR-430 miRNAs rescues the brain defects in MZdicer mutants, revealing essential roles for miRNAs during morphogenesis.


Asunto(s)
Encéfalo/embriología , MicroARNs/fisiología , Morfogénesis , Pez Cebra/embriología , Pez Cebra/genética , Animales , Tipificación del Cuerpo , Diferenciación Celular , Sistema Nervioso Central/embriología , Gástrula/fisiología , Silenciador del Gen , Corazón/embriología , MicroARNs/genética , MicroARNs/metabolismo , Mutación , Neuronas/citología , Fenotipo , Procesamiento Postranscripcional del ARN , ARN Bicatenario/metabolismo , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Transducción de Señal , Somitos/citología , Somitos/fisiología , Médula Espinal/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA