Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 102021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34003109

RESUMEN

Eukaryotic cells use guided search to coordinately control dispersed genetic elements. Argonaute proteins and their small RNA cofactors engage nascent RNAs and chromatin-associated proteins to direct transcriptional silencing. The small ubiquitin-like modifier (SUMO) has been shown to promote the formation and maintenance of silent chromatin (called heterochromatin) in yeast, plants, and animals. Here, we show that Argonaute-directed transcriptional silencing in Caenorhabditis elegans requires SUMOylation of the type 1 histone deacetylase HDA-1. Our findings suggest how SUMOylation promotes the association of HDAC1 with chromatin remodeling factors and with a nuclear Argonaute to initiate de novo heterochromatin silencing.


Asunto(s)
Proteínas Argonautas/genética , Caenorhabditis elegans/genética , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Sumoilación , Transcripción Genética , Animales , Proteínas Argonautas/metabolismo , Ensamble y Desensamble de Cromatina , Silenciador del Gen , Heterocromatina/genética , Heterocromatina/metabolismo , Interferencia de ARN , ARN Interferente Pequeño
2.
Elife ; 102021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34003111

RESUMEN

Germlines shape and balance heredity, integrating and regulating information from both parental and foreign sources. Insights into how germlines handle information have come from the study of factors that specify or maintain the germline fate. In early Caenorhabditis elegans embryos, the CCCH zinc finger protein PIE-1 localizes to the germline where it prevents somatic differentiation programs. Here, we show that PIE-1 also functions in the meiotic ovary where it becomes SUMOylated and engages the small ubiquitin-like modifier (SUMO)-conjugating machinery. Using whole-SUMO-proteome mass spectrometry, we identify HDAC SUMOylation as a target of PIE-1. Our analyses of genetic interactions between pie-1 and SUMO pathway mutants suggest that PIE-1 engages the SUMO machinery both to preserve the germline fate in the embryo and to promote Argonaute-mediated surveillance in the adult germline.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/genética , Sumoilación/genética , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Caenorhabditis elegans/metabolismo , Diferenciación Celular , Femenino , Meiosis , Óvulo , ARN Interferente Pequeño/metabolismo
3.
Mol Cell ; 81(3): 546-557.e5, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33378643

RESUMEN

Eukaryotic cells regulate 5'-triphosphorylated RNAs (ppp-RNAs) to promote cellular functions and prevent recognition by antiviral RNA sensors. For example, RNA capping enzymes possess triphosphatase domains that remove the γ phosphates of ppp-RNAs during RNA capping. Members of the closely related PIR-1 (phosphatase that interacts with RNA and ribonucleoprotein particle 1) family of RNA polyphosphatases remove both the ß and γ phosphates from ppp-RNAs. Here, we show that C. elegans PIR-1 dephosphorylates ppp-RNAs made by cellular RNA-dependent RNA polymerases (RdRPs) and is required for the maturation of 26G-RNAs, Dicer-dependent small RNAs that regulate thousands of genes during spermatogenesis and embryogenesis. PIR-1 also regulates the CSR-1 22G-RNA pathway and has critical functions in both somatic and germline development. Our findings suggest that PIR-1 modulates both Dicer-dependent and Dicer-independent Argonaute pathways and provide insight into how cells and viruses use a conserved RNA phosphatase to regulate and respond to ppp-RNA species.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Monoéster Fosfórico Hidrolasas/metabolismo , Procesamiento Postranscripcional del ARN , ARN/metabolismo , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Regulación del Desarrollo de la Expresión Génica , Monoéster Fosfórico Hidrolasas/genética , Fosforilación , ARN/genética , Caperuzas de ARN , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Espermatogénesis , Especificidad por Sustrato
4.
Gastroenterology ; 152(5): 1161-1173.e1, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27956228

RESUMEN

BACKGROUND & AIMS: It has been a challenge to identify liver tumor suppressors or oncogenes due to the genetic heterogeneity of these tumors. We performed a genome-wide screen to identify suppressors of liver tumor formation in mice, using CRISPR-mediated genome editing. METHODS: We performed a genome-wide CRISPR/Cas9-based knockout screen of P53-null mouse embryonic liver progenitor cells that overexpressed MYC. We infected p53-/-;Myc;Cas9 hepatocytes with the mGeCKOa lentiviral library of 67,000 single-guide RNAs (sgRNAs), targeting 20,611 mouse genes, and transplanted the transduced cells subcutaneously into nude mice. Within 1 month, all the mice that received the sgRNA library developed subcutaneous tumors. We performed high-throughput sequencing of tumor DNA and identified sgRNAs increased at least 8-fold compared to the initial cell pool. To validate the top 10 candidate tumor suppressors from this screen, we collected data from patients with hepatocellular carcinoma (HCC) using the Cancer Genome Atlas and COSMIC databases. We used CRISPR to inactivate candidate tumor suppressor genes in p53-/-;Myc;Cas9 cells and transplanted them subcutaneously into nude mice; tumor formation was monitored and tumors were analyzed by histology and immunohistochemistry. Mice with liver-specific disruption of p53 were given hydrodynamic tail-vein injections of plasmids encoding Myc and sgRNA/Cas9 designed to disrupt candidate tumor suppressors; growth of tumors and metastases was monitored. We compared gene expression profiles of liver cells with vs without tumor suppressor gene disrupted by sgRNA/Cas9. Genes found to be up-regulated after tumor suppressor loss were examined in liver cancer cell lines; their expression was knocked down using small hairpin RNAs, and tumor growth was examined in nude mice. Effects of the MEK inhibitors AZD6244, U0126, and trametinib, or the multi-kinase inhibitor sorafenib, were examined in human and mouse HCC cell lines. RESULTS: We identified 4 candidate liver tumor suppressor genes not previously associated with liver cancer (Nf1, Plxnb1, Flrt2, and B9d1). CRISPR-mediated knockout of Nf1, a negative regulator of RAS, accelerated liver tumor formation in mice. Loss of Nf1 or activation of RAS up-regulated the liver progenitor cell markers HMGA2 and SOX9. RAS pathway inhibitors suppressed the activation of the Hmga2 and Sox9 genes that resulted from loss of Nf1 or oncogenic activation of RAS. Knockdown of HMGA2 delayed formation of xenograft tumors from cells that expressed oncogenic RAS. In human HCCs, low levels of NF1 messenger RNA or high levels of HMGA2 messenger RNA were associated with shorter patient survival time. Liver cancer cells with inactivation of Plxnb1, Flrt2, and B9d1 formed more tumors in mice and had increased levels of mitogen-activated protein kinase phosphorylation. CONCLUSIONS: Using a CRISPR-based strategy, we identified Nf1, Plxnb1, Flrt2, and B9d1 as suppressors of liver tumor formation. We validated the observation that RAS signaling, via mitogen-activated protein kinase, contributes to formation of liver tumors in mice. We associated decreased levels of NF1 and increased levels of its downstream protein HMGA2 with survival times of patients with HCC. Strategies to inhibit or reduce HMGA2 might be developed to treat patients with liver cancer.


Asunto(s)
Carcinoma Hepatocelular/genética , Regulación Neoplásica de la Expresión Génica , Hepatocitos/metabolismo , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteína p53 Supresora de Tumor/genética , Animales , Bencimidazoles/farmacología , Western Blotting , Butadienos/farmacología , Sistemas CRISPR-Cas , Línea Celular Tumoral , Proteínas del Citoesqueleto , ADN de Neoplasias/genética , Inhibidores Enzimáticos , Genes de Neurofibromatosis 1 , Estudio de Asociación del Genoma Completo , Proteínas HMGA/genética , Proteína HMGA2/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunohistoquímica , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Ratones Desnudos , Proteínas del Tejido Nervioso/genética , Niacinamida/análogos & derivados , Niacinamida/farmacología , Nitrilos/farmacología , Compuestos de Fenilurea/farmacología , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Piridonas/farmacología , Pirimidinonas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/genética , Análisis de Secuencia de ADN , Sorafenib , Análisis de Supervivencia , Proteínas Supresoras de Tumor/genética , Proteínas ras/genética
5.
Curr Protoc Mol Biol ; 109: 26.3.1-26.3.30, 2015 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-25559107

RESUMEN

RNAi has become an essential tool in C. elegans research. This unit describes procedures for RNAi in C. elegans by microinjecting with dsRNA, feeding with bacteria expressing dsRNA, and soaking in dsRNA solution, as well as high-throughput methods for RNAi-based screens.


Asunto(s)
Caenorhabditis elegans/genética , Biología Molecular/métodos , Parasitología/métodos , Interferencia de ARN , Animales , Pruebas Genéticas/métodos
6.
Cell ; 160(3): 407-19, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25635455

RESUMEN

Effective silencing by RNA-interference (RNAi) depends on mechanisms that amplify and propagate the silencing signal. In some organisms, small-interfering RNAs (siRNAs) are amplified from target mRNAs by RNA-dependent RNA polymerase (RdRP). Both RdRP recruitment and mRNA silencing require Argonaute proteins, which are generally thought to degrade RNAi targets by directly cleaving them. However, in C. elegans, the enzymatic activity of the primary Argonaute, RDE-1, is not required for silencing activity. We show that RDE-1 can instead recruit an endoribonuclease, RDE-8, to target RNA. RDE-8 can cleave RNA in vitro and is needed for the production of 3' uridylated fragments of target mRNA in vivo. We also find that RDE-8 promotes RdRP activity, thereby ensuring amplification of siRNAs. Together, our findings suggest a model in which RDE-8 cleaves target mRNAs to mediate silencing, while generating 3' uridylated mRNA fragments to serve as templates for the RdRP-directed amplification of the silencing signal.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Endorribonucleasas/metabolismo , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Gránulos Citoplasmáticos/metabolismo , Endorribonucleasas/química , Endorribonucleasas/genética , Datos de Secuencia Molecular , Interferencia de ARN , ARN Bicatenario , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleasa III/metabolismo , Alineación de Secuencia
7.
Elife ; 32014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-24966209

RESUMEN

Virus-host interactions drive a remarkable diversity of immune responses and countermeasures. We found that two RNA viruses with broad host ranges, vesicular stomatitis virus (VSV) and Sindbis virus (SINV), are completely restricted in their replication after entry into Lepidopteran cells. This restriction is overcome when cells are co-infected with vaccinia virus (VACV), a vertebrate DNA virus. Using RNAi screening, we show that Lepidopteran RNAi, Nuclear Factor-κB, and ubiquitin-proteasome pathways restrict RNA virus infection. Surprisingly, a highly conserved, uncharacterized VACV protein, A51R, can partially overcome this virus restriction. We show that A51R is also critical for VACV replication in vertebrate cells and for pathogenesis in mice. Interestingly, A51R colocalizes with, and stabilizes, host microtubules and also associates with ubiquitin. We show that A51R promotes viral protein stability, possibly by preventing ubiquitin-dependent targeting of viral proteins for destruction. Importantly, our studies reveal exciting new opportunities to study virus-host interactions in experimentally-tractable Lepidopteran systems.


Asunto(s)
Lepidópteros/virología , Virus Vaccinia/fisiología , Vesiculovirus/fisiología , Proteínas Virales/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , ADN Viral/química , Humanos , Lepidópteros/inmunología , Ratones , Microscopía Confocal , Microscopía Fluorescente , Complejo de la Endopetidasa Proteasomal/química , Interferencia de ARN , Ubiquitina/química , Replicación Viral
8.
Genetics ; 197(4): 1069-80, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24879462

RESUMEN

Genome editing based on CRISPR (clustered regularly interspaced short palindromic repeats)-associated nuclease (Cas9) has been successfully applied in dozens of diverse plant and animal species, including the nematode Caenorhabditis elegans. The rapid life cycle and easy access to the ovary by micro-injection make C. elegans an ideal organism both for applying CRISPR-Cas9 genome editing technology and for optimizing genome-editing protocols. Here we report efficient and straightforward CRISPR-Cas9 genome-editing methods for C. elegans, including a Co-CRISPR strategy that facilitates detection of genome-editing events. We describe methods for detecting homologous recombination (HR) events, including direct screening methods as well as new selection/counterselection strategies. Our findings reveal a surprisingly high frequency of HR-mediated gene conversion, making it possible to rapidly and precisely edit the C. elegans genome both with and without the use of co-inserted marker genes.


Asunto(s)
Proteínas Asociadas a CRISPR/genética , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Desoxirribonucleasas/genética , Genoma de los Helmintos , Animales , Secuencia de Bases , Marcadores Genéticos , Recombinación Homóloga/genética , Datos de Secuencia Molecular , Mutagénesis Insercional , Plásmidos/genética , Análisis de Secuencia de ADN
9.
Longev Healthspan ; 3: 5, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24834345

RESUMEN

BACKGROUND: Insulin/IGF-1 signaling plays a central role in longevity across phylogeny. In C. elegans, the forkhead box O (FOXO) transcription factor, DAF-16, is the primary target of insulin/IGF-1 signaling, and multiple isoforms of DAF-16 (a, b, and d/f) modulate lifespan, metabolism, dauer formation, and stress resistance. Thus far, across phylogeny modulation of mammalian FOXOs and DAF-16 have focused on post-translational regulation with little focus on transcriptional regulation. In C. elegans, we have previously shown that DAF-16d/f cooperates with DAF-16a to promote longevity. In this study, we generated transgenic strains expressing near-endogenous levels of either daf-16a or daf-16d/f, and examined temporal expression of the isoforms to further define how these isoforms contribute to lifespan regulation. RESULTS: Here, we show that DAF-16a is sensitive both to changes in gene dosage and to alterations in the level of insulin/IGF-1 signaling. Interestingly, we find that as worms age, the intestinal expression of daf-16d/f but not daf-16a is dramatically upregulated at the level of transcription. Preventing this transcriptional upregulation shortens lifespan, indicating that transcriptional regulation of daf-16d/f promotes longevity. In an RNAi screen of transcriptional regulators, we identify elt-2 (GATA transcription factor) and swsn-1 (core subunit of SWI/SNF complex) as key modulators of daf-16d/f gene expression. ELT-2 and another GATA factor, ELT-4, promote longevity via both DAF-16a and DAF-16d/f while the components of SWI/SNF complex promote longevity specifically via DAF-16d/f. CONCLUSIONS: Our findings indicate that transcriptional control of C. elegans FOXO/daf-16 is an essential regulatory event. Considering the conservation of FOXO across species, our findings identify a new layer of FOXO regulation as a potential determinant of mammalian longevity and age-related diseases such as cancer and diabetes.

10.
Elife ; 3: e01460, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24550252

RESUMEN

Oncogenic mutations in BRAF and NRAS occur in 70% of melanomas. In this study, we identify a microRNA, miR-146a, that is highly upregulated by oncogenic BRAF and NRAS. Expression of miR-146a increases the ability of human melanoma cells to proliferate in culture and form tumors in mice, whereas knockdown of miR-146a has the opposite effects. We show these oncogenic activities are due to miR-146a targeting the NUMB mRNA, a repressor of Notch signaling. Previous studies have shown that pre-miR-146a contains a single nucleotide polymorphism (C>G rs2910164). We find that the ability of pre-miR-146a/G to activate Notch signaling and promote oncogenesis is substantially higher than that of pre-miR-146a/C. Analysis of melanoma cell lines and matched patient samples indicates that during melanoma progression pre-miR-146a/G is enriched relative to pre-miR-146a/C, resulting from a C-to-G somatic mutation in pre-miR-146a/C. Collectively, our results reveal a central role for miR-146a in the initiation and progression of melanoma. DOI: http://dx.doi.org/10.7554/eLife.01460.001.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Melanoma/metabolismo , MicroARNs/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/secundario , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Desnudos , MicroARNs/genética , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Receptores Notch/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Factores de Tiempo , Transfección , Carga Tumoral
11.
Cell ; 155(7): 1532-44, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24360276

RESUMEN

During each life cycle, germ cells preserve and pass on both genetic and epigenetic information. In C. elegans, the ALG-3/4 Argonaute proteins are expressed during male gametogenesis and promote male fertility. Here, we show that the CSR-1 Argonaute functions with ALG-3/4 to positively regulate target genes required for spermiogenesis. Our findings suggest that ALG-3/4 functions during spermatogenesis to amplify a small RNA signal that represents an epigenetic memory of male-specific gene expression. CSR-1, which is abundant in mature sperm, appears to transmit this memory to offspring. Surprisingly, in addition to small RNAs targeting male-specific genes, we show that males also harbor an extensive repertoire of CSR-1 small RNAs targeting oogenesis-specific mRNAs. Together, these findings suggest that C. elegans sperm transmit not only the genome but also epigenetic binary signals in the form of Argonaute/small RNA complexes that constitute a memory of gene expression in preceding generations.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Epigénesis Genética , Proteínas de Unión al ARN/metabolismo , Espermatogénesis , Animales , Caenorhabditis elegans/genética , Femenino , Masculino , ARN Pequeño no Traducido/metabolismo , Transducción de Señal , Espermatozoides , Transcripción Genética
12.
Dev Cell ; 27(6): 656-63, 2013 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-24360782

RESUMEN

Organisms can develop adaptive sequence-specific immunity by reexpressing pathogen-specific small RNAs that guide gene silencing. For example, the C. elegans PIWI-Argonaute/piwi-interacting RNA (piRNA) pathway recruits RNA-dependent RNA polymerase (RdRP) to foreign sequences to amplify a transgenerational small-RNA-induced epigenetic silencing signal (termed RNAe). Here, we provide evidence that, in addition to an adaptive memory of silenced sequences, C. elegans can also develop an opposing adaptive memory of expressed/self-mRNAs. We refer to this mechanism, which can prevent or reverse RNAe, as RNA-induced epigenetic gene activation (RNAa). We show that CSR-1, which engages RdRP-amplified small RNAs complementary to germline-expressed mRNAs, is required for RNAa. We show that a transgene with RNAa activity also exhibits accumulation of cognate CSR-1 small RNAs. Our findings suggest that C. elegans adaptively acquires and maintains a transgenerational CSR-1 memory that recognizes and protects self-mRNAs, allowing piRNAs to recognize foreign sequences innately, without the need for prior exposure


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Epigénesis Genética , Silenciador del Gen , Células Germinativas/metabolismo , ARN de Helminto/genética , ARN Interferente Pequeño/metabolismo , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , ARN de Helminto/metabolismo , ARN Interferente Pequeño/genética , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Transducción de Señal
13.
Proc Natl Acad Sci U S A ; 110(10): E918-27, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23431196

RESUMEN

In early Caenorhabditis elegans embryos, the Wingless/int (Wnt)- and Src-signaling pathways function in parallel to induce both the division orientation of the endomesoderm (EMS) blastomere and the endoderm fate of the posterior EMS daughter cell, called E. Here, we show that, in addition to its role in endoderm specification, the ß-catenin-related protein Worm armadillo 1 (WRM-1) also plays a role in controlling EMS division orientation. WRM-1 localizes to the cortex of cells in both embryos and larvae and is released from the cortex in a Wnt-responsive manner. We show that WRM-1 cortical release is disrupted in a hypomorphic cyclin-dependent protein kinase 1 (cdk-1) mutant and that WRM-1 lacking potential CDK-1 phosphoacceptor sites is retained at the cortex. In both cases, cortical WRM-1 interferes with EMS spindle rotation without affecting endoderm specification. Finally, we show that removal of WRM-1 from the cortex can restore WT division orientation, even when both Wnt- and Src-signaling pathways are compromised. Our findings are consistent with a model in which Wnt signaling and CDK-1 modify WRM-1 in a temporal and spatial manner to unmask an intrinsic polarity cue required for proper orientation of the EMS cell division axis.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , División Celular/genética , División Celular/fisiología , Polaridad Celular/genética , Polaridad Celular/fisiología , Genes de Helminto , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Profase/genética , Profase/fisiología , Homología de Secuencia de Aminoácido , Transducción de Señal , Huso Acromático/metabolismo , Vía de Señalización Wnt , Familia-src Quinasas/metabolismo
14.
Cell ; 151(7): 1488-500, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23260138

RESUMEN

Piwi-interacting (pi) RNAs are germline-expressed small RNAs linked to epigenetic programming. C. elegans piRNAs are thought to be transcribed as independent gene-like loci. To test this idea and to identify potential transcription start (TS) sites for piRNA precursors, we developed CapSeq, an efficient enzymatic method for 5' anchored RNA profiling. Using CapSeq, we identify candidate TS sites, defined by 70-90 nt sequence tags, for >50% of annotated Pol II loci. Surprisingly, however, these CapSeq tags failed to identify the overwhelming majority of piRNA loci. Instead, we show that the likely piRNA precursors are ∼26 nt capped small (cs) RNAs that initiate precisely 2 nt upstream of mature piRNAs and that piRNA processing or stability requires a U at the csRNA +3 position. Finally, we identify a heretofore unrecognized class of piRNAs processed from csRNAs that are expressed at promoters genome wide, nearly doubling the number of piRNAs available for genome surveillance.


Asunto(s)
Caenorhabditis elegans/genética , Perfilación de la Expresión Génica/métodos , Caperuzas de ARN/genética , Procesamiento Postranscripcional del ARN , ARN de Helminto/genética , ARN Interferente Pequeño/genética , Sitio de Iniciación de la Transcripción , Animales , Proteínas Argonautas/metabolismo , Caenorhabditis elegans/metabolismo , Estudio de Asociación del Genoma Completo , Ratones , Motivos de Nucleótidos , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo
15.
Cell ; 150(1): 78-87, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22738724

RESUMEN

Piwi Argonautes and Piwi-interacting RNAs (piRNAs) mediate genome defense by targeting transposons. However, many piRNA species lack obvious sequence complementarity to transposons or other loci; only one C. elegans transposon is a known piRNA target. Here, we show that, in mutants lacking the Piwi Argonaute PRG-1 (and consequently its associated piRNAs/21U-RNAs), many silent loci in the germline exhibit increased levels of mRNA expression with a concomitant depletion of RNA-dependent RNA polymerase (RdRP)-derived secondary small RNAs termed 22G-RNAs. Sequences depleted of 22G-RNAs are proximal to potential target sites that base pair imperfectly but extensively to 21U-RNAs. We show that PRG-1 is required to initiate, but not to maintain, silencing of transgenes engineered to contain complementarity to endogenous 21U-RNAs. Our findings support a model in which C. elegans piRNAs utilize their enormous repertoire of targeting capacity to scan the germline transcriptome for foreign sequences, while endogenous germline-expressed genes are actively protected from piRNA-induced silencing.


Asunto(s)
Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Genoma de los Helmintos , ARN de Helminto/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Proteínas Argonautas/metabolismo , Silenciador del Gen , Células Germinativas
16.
Cell ; 150(1): 65-77, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22738726

RESUMEN

Organisms employ a fascinating array of strategies to silence invasive nucleic acids such as transposons and viruses. Although evidence exists for several pathways that detect foreign sequences, including pathways that sense copy number, unpaired DNA, or aberrant RNA (e.g., dsRNA), in many cases, the mechanisms used to distinguish "self" from "nonself" nucleic acids remain mysterious. Here, we describe an RNA-induced epigenetic silencing pathway that permanently silences single-copy transgenes. We show that the Piwi Argonaute PRG-1 and its genomically encoded piRNA cofactors initiate permanent silencing, and maintenance depends on chromatin factors and the WAGO Argonaute pathway. Our findings support a model in which PRG-1 scans for foreign sequences and two other Argonaute pathways serve as epigenetic memories of "self" and "nonself" RNAs. These findings suggest how organisms can utilize RNAi-related mechanisms to detect foreign sequences not by any molecular signature, but by comparing the foreign sequence to a memory of previous gene expression.


Asunto(s)
Caenorhabditis elegans/genética , Epigenómica , ARN de Helminto/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Silenciador del Gen , Células Germinativas/metabolismo , Interferencia de ARN
17.
Nat Struct Mol Biol ; 19(1): 90-7, 2011 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-22179787

RESUMEN

Endogenous RNA interference (endo-RNAi) pathways use a variety of mechanisms to generate siRNA and to mediate gene silencing. In Caenorhabditis elegans, DCR-1 is essential for competing RNAi pathways-the ERI endo-RNAi pathway and the exogenous RNAi pathway-to function. Here, we demonstrate that DCR-1 forms exclusive complexes in each pathway and further define the ERI-DCR-1 complex. We show that the tandem tudor protein ERI-5 potentiates ERI endo-RNAi by tethering an RNA-dependent RNA polymerase (RdRP) module to DCR-1. In the absence of ERI-5, the RdRP module is uncoupled from DCR-1. Notably, EKL-1, an ERI-5 paralog that specifies distinct RdRP modules in Dicer-independent endo-RNAi pathways, partially compensates for the loss of ERI-5 without interacting with DCR-1. Our results implicate tudor proteins in the recruitment of RdRP complexes to specific steps within DCR-1-dependent and DCR-1-independent endo-RNAi pathways.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Portadoras/metabolismo , Interferencia de ARN , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleasa III/metabolismo , Animales , Western Blotting , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas Portadoras/genética , Embrión no Mamífero/metabolismo , Inmunoprecipitación , Mutación , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Polimerasa Dependiente del ARN/genética , Complejo Silenciador Inducido por ARN/genética , Complejo Silenciador Inducido por ARN/metabolismo , Proteínas Recombinantes/metabolismo , Ribonucleasa III/genética
18.
Methods Mol Biol ; 725: 251-80, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21528459

RESUMEN

Small RNA pathways fulfill a plethora of gene-regulatory functions in a variety of organisms. In the nematode worm, Caenorhabditis elegans, a number of endogenous small RNA pathways have been described, including the microRNA pathway, the 21U/piRNA pathway, the 26G-RNA pathways, and the 22G-RNA pathways. Argonaute proteins are key effector molecules of each pathway that, together with their small RNA cofactors regulate various processes including developmental timing, fertility, transposon silencing, and chromosome segregation. Although several of the 26 Argonautes in the worm have been studied to date, a number have yet to be fully characterized or their small RNA binding complement defined. The identification of small RNAs that copurify with an Argonaute family member is central to understanding the targets and assessing the function of that Argonaute. Here we discuss the rationale for generating reagents to immunoprecipitate Argonaute complexes and provide a cohesive protocol for the cloning and Illumina deep-sequencing of Argonaute-associated small RNAs in C. elegans.


Asunto(s)
Caenorhabditis elegans/genética , Clonación Molecular , Factores Eucarióticos de Iniciación/metabolismo , MicroARNs/genética , Animales , Anticuerpos/inmunología , Anticuerpos/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Epítopos/inmunología , Epítopos/metabolismo , Factores Eucarióticos de Iniciación/inmunología , Biblioteca de Genes , Inmunoprecipitación , MicroARNs/aislamiento & purificación , MicroARNs/metabolismo , Control de Calidad , Reproducibilidad de los Resultados
19.
Cell ; 140(4): 452-4, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20178736

RESUMEN

The amplification of small RNAs and the assembly of heterochromatin are mutually dependent processes in fission yeast. But which comes first? Halic and Moazed (2010) propose that primal small RNAs initiate the amplification of small interfering RNAs that drive heterochromatin formation and chromatin silencing.


Asunto(s)
Heterocromatina/metabolismo , Interferencia de ARN , ARN no Traducido/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , ARN Interferente Pequeño/metabolismo
20.
Proc Natl Acad Sci U S A ; 107(8): 3582-7, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20133583

RESUMEN

Argonaute (AGO) proteins interact with distinct classes of small RNAs to direct multiple regulatory outcomes. In many organisms, including plants, fungi, and nematodes, cellular RNA-dependent RNA polymerases (RdRPs) use AGO targets as templates for amplification of silencing signals. Here, we show that distinct RdRPs function sequentially to produce small RNAs that target endogenous loci in Caenorhabditis elegans. We show that DCR-1, the RdRP RRF-3, and the dsRNA-binding protein RDE-4 are required for the biogenesis of 26-nt small RNAs with a 5' guanine (26G-RNAs) and that 26G-RNAs engage the Piwi-clade AGO, ERGO-1. Our findings support a model in which targeting by ERGO-1 recruits a second RdRP (RRF-1 or EGO-1), which in turn transcribes 22G-RNAs that interact with worm-specific AGOs (WAGOs) to direct gene silencing. ERGO-1 targets exhibit a nonrandom distribution in the genome and appear to include many gene duplications, suggesting that this pathway may control overexpression resulting from gene expansion.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/biosíntesis , Proteínas de Unión al ARN/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Endorribonucleasas/metabolismo , Silenciador del Gen , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Ribonucleasa III , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...