Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(36): e2303859120, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37639593

RESUMEN

Recurrent chromosomal rearrangements found in rhabdomyosarcoma (RMS) produce the PAX3-FOXO1 fusion protein, which is an oncogenic driver and a dependency in this disease. One important function of PAX3-FOXO1 is to arrest myogenic differentiation, which is linked to the ability of RMS cells to gain an unlimited proliferation potential. Here, we developed a phenotypic screening strategy for identifying factors that collaborate with PAX3-FOXO1 to block myo-differentiation in RMS. Unlike most genes evaluated in our screen, we found that loss of any of the three subunits of the Nuclear Factor Y (NF-Y) complex leads to a myo-differentiation phenotype that resembles the effect of inactivating PAX3-FOXO1. While the transcriptomes of NF-Y- and PAX3-FOXO1-deficient RMS cells bear remarkable similarity to one another, we found that these two transcription factors occupy nonoverlapping sites along the genome: NF-Y preferentially occupies promoters, whereas PAX3-FOXO1 primarily binds to distal enhancers. By integrating multiple functional approaches, we map the PAX3 promoter as the point of intersection between these two regulators. We show that NF-Y occupies CCAAT motifs present upstream of PAX3 to function as a transcriptional activator of PAX3-FOXO1 expression in RMS. These findings reveal a critical upstream role of NF-Y in the oncogenic PAX3-FOXO1 pathway, highlighting how a broadly essential transcription factor can perform tumor-specific roles in governing cellular state.


Asunto(s)
Rabdomiosarcoma , Factor de Unión a CCAAT/genética , Diferenciación Celular/genética , Aberraciones Cromosómicas , Rabdomiosarcoma/genética , Factores de Transcripción
2.
Nature ; 607(7917): 169-175, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35576971

RESUMEN

Tuft cells are a rare chemosensory lineage that coordinates immune and neural responses to foreign pathogens in mucosal tissues1. Recent studies have also revealed tuft-cell-like human tumours2,3, particularly as a variant of small-cell lung cancer. Both normal and neoplastic tuft cells share a genetic requirement for the transcription factor POU2F3 (refs. 2,4), although the transcriptional mechanisms that generate this cell type are poorly understood. Here we show that binding of POU2F3 to the uncharacterized proteins C11orf53 and COLCA2 (renamed here OCA-T1/POU2AF2 and OCA-T2/POU2AF3, respectively) is critical in the tuft cell lineage. OCA-T1 and OCA-T2 are paralogues of the B-cell-specific coactivator OCA-B; all three proteins are encoded in a gene cluster and contain a conserved peptide that binds to class II POU transcription factors and a DNA octamer motif in a bivalent manner. We demonstrate that binding between POU2F3 and OCA-T1 or OCA-T2 is essential in tuft-cell-like small-cell lung cancer. Moreover, we generated OCA-T1-deficient mice, which are viable but lack tuft cells in several mucosal tissues. These findings reveal that the POU2F3-OCA-T complex is the master regulator of tuft cell identity and a molecular vulnerability of tuft-cell-like small-cell lung cancer.


Asunto(s)
Linaje de la Célula , Neoplasias Pulmonares , Proteínas de Neoplasias , Factores de Transcripción de Octámeros , Carcinoma Pulmonar de Células Pequeñas , Animales , Humanos , Ratones , Neoplasias Pulmonares/patología , Membrana Mucosa/patología , Familia de Multigenes/genética , Proteínas de Neoplasias/metabolismo , Motivos de Nucleótidos , Factores de Transcripción de Octámeros/metabolismo , Factores del Dominio POU/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Transactivadores
3.
Gut ; 71(5): 879-888, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35144974

RESUMEN

OBJECTIVE: We assessed whether famotidine improved inflammation and symptomatic recovery in outpatients with mild to moderate COVID-19. DESIGN: Randomised, double-blind, placebo-controlled, fully remote, phase 2 clinical trial (NCT04724720) enrolling symptomatic unvaccinated adult outpatients with confirmed COVID-19 between January 2021 and April 2021 from two US centres. Patients self-administered 80 mg famotidine (n=28) or placebo (n=27) orally three times a day for 14 consecutive days. Endpoints were time to (primary) or rate of (secondary) symptom resolution, and resolution of inflammation (exploratory). RESULTS: Of 55 patients in the intention-to-treat group (median age 35 years (IQR: 20); 35 women (64%); 18 African American (33%); 14 Hispanic (26%)), 52 (95%) completed the trial, submitting 1358 electronic symptom surveys. Time to symptom resolution was not statistically improved (p=0.4). Rate of symptom resolution was improved for patients taking famotidine (p<0.0001). Estimated 50% reduction of overall baseline symptom scores were achieved at 8.2 days (95% CI: 7 to 9.8 days) for famotidine and 11.4 days (95% CI: 10.3 to 12.6 days) for placebo treated patients. Differences were independent of patient sex, race or ethnicity. Five self-limiting adverse events occurred (famotidine, n=2 (40%); placebo, n=3 (60%)). On day 7, fewer patients on famotidine had detectable interferon alpha plasma levels (p=0.04). Plasma immunoglobulin type G levels to SARS-CoV-2 nucleocapsid core protein were similar between both arms. CONCLUSIONS: Famotidine was safe and well tolerated in outpatients with mild to moderate COVID-19. Famotidine led to earlier resolution of symptoms and inflammation without reducing anti-SARS-CoV-2 immunity. Additional randomised trials are required.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Famotidina , Adulto , Método Doble Ciego , Famotidina/uso terapéutico , Femenino , Humanos , Inflamación , SARS-CoV-2 , Resultado del Tratamiento
4.
Clin Cancer Res ; 27(7): 2023-2037, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33495315

RESUMEN

PURPOSE: Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease characterized by an extensive fibroinflammatory stroma, which includes abundant cancer-associated fibroblast (CAF) populations. PDAC CAFs are heterogeneous, but the nature of this heterogeneity is incompletely understood. The Hedgehog pathway functions in PDAC in a paracrine manner, with ligands secreted by cancer cells signaling to stromal cells in the microenvironment. Previous reports investigating the role of Hedgehog signaling in PDAC have been contradictory, with Hedgehog signaling alternately proposed to promote or restrict tumor growth. In light of the newly discovered CAF heterogeneity, we investigated how Hedgehog pathway inhibition reprograms the PDAC microenvironment. EXPERIMENTAL DESIGN: We used a combination of pharmacologic inhibition, gain- and loss-of-function genetic experiments, cytometry by time-of-flight, and single-cell RNA sequencing to study the roles of Hedgehog signaling in PDAC. RESULTS: We found that Hedgehog signaling is uniquely activated in fibroblasts and differentially elevated in myofibroblastic CAFs (myCAF) compared with inflammatory CAFs (iCAF). Sonic Hedgehog overexpression promotes tumor growth, while Hedgehog pathway inhibition with the smoothened antagonist, LDE225, impairs tumor growth. Furthermore, Hedgehog pathway inhibition reduces myCAF numbers and increases iCAF numbers, which correlates with a decrease in cytotoxic T cells and an expansion in regulatory T cells, consistent with increased immunosuppression. CONCLUSIONS: Hedgehog pathway inhibition alters fibroblast composition and immune infiltration in the pancreatic cancer microenvironment.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal Pancreático/patología , Proteínas Hedgehog/fisiología , Neoplasias Pancreáticas/patología , Animales , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/inmunología , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Transducción de Señal/fisiología , Microambiente Tumoral
5.
Proc Natl Acad Sci U S A ; 117(21): 11471-11482, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32385160

RESUMEN

Lineage plasticity is a prominent feature of pancreatic ductal adenocarcinoma (PDA) cells, which can occur via deregulation of lineage-specifying transcription factors. Here, we show that the zinc finger protein ZBED2 is aberrantly expressed in PDA and alters tumor cell identity in this disease. Unexpectedly, our epigenomic experiments reveal that ZBED2 is a sequence-specific transcriptional repressor of IFN-stimulated genes, which occurs through antagonism of IFN regulatory factor 1 (IRF1)-mediated transcriptional activation at cooccupied promoter elements. Consequently, ZBED2 attenuates the transcriptional output and growth arrest phenotypes downstream of IFN signaling in multiple PDA cell line models. We also found that ZBED2 is preferentially expressed in the squamous molecular subtype of human PDA, in association with inferior patient survival outcomes. Consistent with this observation, we show that ZBED2 can repress the pancreatic progenitor transcriptional program, enhance motility, and promote invasion in PDA cells. Collectively, our findings suggest that high ZBED2 expression is acquired during PDA progression to suppress the IFN response pathway and to promote lineage plasticity in this disease.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Proteínas de Unión al ADN/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Neoplasias Pancreáticas/patología , Factores de Transcripción/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/mortalidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Factor 1 Regulador del Interferón/genética , Interferón gamma/farmacología , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Regiones Promotoras Genéticas , Análisis de Supervivencia , Factores de Transcripción/genética
6.
Elife ; 92020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32329713

RESUMEN

A highly aggressive subset of pancreatic ductal adenocarcinomas undergo trans-differentiation into the squamous lineage during disease progression. Here, we investigated whether squamous trans-differentiation of human and mouse pancreatic cancer cells can influence the phenotype of non-neoplastic cells in the tumor microenvironment. Conditioned media experiments revealed that squamous pancreatic cancer cells secrete factors that recruit neutrophils and convert pancreatic stellate cells into cancer-associated fibroblasts (CAFs) that express inflammatory cytokines at high levels. We use gain- and loss-of-function approaches to show that squamous-subtype pancreatic tumor models become enriched with neutrophils and inflammatory CAFs in a p63-dependent manner. These effects occur, at least in part, through p63-mediated activation of enhancers at pro-inflammatory cytokine loci, which includes IL1A and CXCL1 as key targets. Taken together, our findings reveal enhanced tissue inflammation as a consequence of squamous trans-differentiation in pancreatic cancer, thus highlighting an instructive role of tumor cell lineage in reprogramming the stromal microenvironment.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Transdiferenciación Celular/fisiología , Inflamación/patología , Neoplasias Pancreáticas/patología , Animales , Fibroblastos Asociados al Cáncer/fisiología , Carcinoma Ductal Pancreático/inmunología , Linaje de la Célula , Citocinas/genética , Citocinas/fisiología , Humanos , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Neoplasias Pancreáticas/inmunología , Células del Estroma/patología , Microambiente Tumoral
7.
Cell Syst ; 9(5): 475-482.e4, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31677971

RESUMEN

A small population of αß T cells is characterized by the expression of more than one unique T cell receptor (TCR); this outcome is the result of "allelic inclusion," that is, inclusion of both α- or ß-chain alleles during V(D)J recombination. Limitations in single-cell sequencing technology, however, have precluded comprehensive enumeration of these dual receptor T cells. Here, we develop and experimentally validate a fully Bayesian inference model capable of reliably estimating the true rate of α and ß TCR allelic inclusion across two different emulsion-barcoding single-cell sequencing platforms. We provide a database composed of over 51,000 previously unpublished allelic inclusion TCR sequence sets drawn from eight healthy individuals and show that allelic inclusion contributes a distinct and functionally important set of sequences to the human TCR repertoire. This database and a Python implementation of our statistical inference model are freely available at our Github repository (https://github.com/JasonACarter/Allelic_inclusion).


Asunto(s)
Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T/genética , Alelos , Teorema de Bayes , Bases de Datos Genéticas , Frecuencia de los Genes/genética , Humanos , Receptores de Antígenos de Linfocitos T/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Análisis de la Célula Individual/métodos , Linfocitos T/metabolismo
8.
Genes Dev ; 33(17-18): 1208-1220, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31416967

RESUMEN

The PIWI-interacting RNA (piRNA) pathway is a conserved small RNA-based immune system that protects animal germ cell genomes from the harmful effects of transposon mobilization. In Drosophila ovaries, most piRNAs originate from dual-strand clusters, which generate piRNAs from both genomic strands. Dual-strand clusters use noncanonical transcription mechanisms. Although transcribed by RNA polymerase II, cluster transcripts lack splicing signatures and poly(A) tails. mRNA processing is important for general mRNA export mediated by nuclear export factor 1 (Nxf1). Although UAP56, a component of the transcription and export complex, has been implicated in piRNA precursor export, it remains unknown how dual-strand cluster transcripts are specifically targeted for piRNA biogenesis by export from the nucleus to cytoplasmic processing centers. Here we report that dual-strand cluster transcript export requires CG13741/Bootlegger and the Drosophila nuclear export factor family protein Nxf3. Bootlegger is specifically recruited to piRNA clusters and in turn brings Nxf3. We found that Nxf3 specifically binds to piRNA precursors and is essential for their export to piRNA biogenesis sites, a process that is critical for germline transposon silencing. Our data shed light on how dual-strand clusters compensate for a lack of canonical features of mature mRNAs to be specifically exported via Nxf3, ensuring proper piRNA production.


Asunto(s)
Transporte Activo de Núcleo Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Precursores del ARN/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Elementos Transponibles de ADN/genética , Drosophila/genética , Proteínas de Drosophila/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Unión al ARN/genética
9.
Front Immunol ; 10: 1516, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417541

RESUMEN

Although structural studies of individual T cell receptors (TCRs) have revealed important roles for both the α and ß chain in directing MHC and antigen recognition, repertoire-level immunogenomic analyses have historically examined the ß chain alone. To determine the amount of useful information about TCR repertoire function encoded within αß pairings, we analyzed paired TCR sequences from nearly 100,000 unique CD4+ and CD8+ T cells captured using two different high-throughput, single-cell sequencing approaches. Our results demonstrate little overlap in the healthy CD4+ and CD8+ repertoires, with shared TCR sequences possessing significantly shorter CDR3 sequences corresponding to higher generation probabilities. We further utilized tools from information theory and machine learning to show that while α and ß chains are only weakly associated with lineage, αß pairings appear to synergistically drive TCR-MHC interactions. Vαß gene pairings were found to be the TCR feature most informative of T cell lineage, supporting the existence of germline-encoded paired αß TCR-MHC interaction motifs. Finally, annotating our TCR pairs using a database of sequences with known antigen specificities, we demonstrate that approximately a third of the T cells possess α and ß chains that each recognize different known antigens, suggesting that αß pairing is critical for the accurate inference of repertoire functionality. Together, these findings provide biological insight into the functional implications of αß pairing and highlight the utility of single-cell sequencing in immunogenomics.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Regiones Determinantes de Complementariedad , Aprendizaje Automático , Receptores de Antígenos de Linfocitos T alfa-beta , Análisis de Secuencia de Proteína , Antígenos/genética , Antígenos/inmunología , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/inmunología , Humanos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología
10.
Cell Stem Cell ; 18(2): 276-90, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26669894

RESUMEN

Complex regulatory networks regulate stem cell behavior and contributions to tissue growth, repair, and homeostasis. A full understanding of the networks controlling stem cell self-renewal and differentiation, however, has not yet been realized. To systematically dissect these networks and identify their components, we performed an unbiased, transcriptome-wide in vivo RNAi screen in female Drosophila germline stem cells (GSCs). Based on characterized cellular defects, we classified 646 identified genes into phenotypic and functional groups and unveiled a comprehensive set of networks regulating GSC maintenance, survival, and differentiation. This analysis revealed an unexpected role for ribosomal assembly factors in controlling stem cell cytokinesis. Moreover, our data show that the transition from self-renewal to differentiation relies on enhanced ribosome biogenesis accompanied by increased protein synthesis. Collectively, these results detail the extensive genetic networks that control stem cell homeostasis and highlight the intricate regulation of protein synthesis during differentiation.


Asunto(s)
Diferenciación Celular , Drosophila melanogaster/citología , Células Germinativas/citología , Biogénesis de Organelos , Biosíntesis de Proteínas , Ribosomas/metabolismo , Células Madre/citología , Animales , Nucléolo Celular/patología , Supervivencia Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes de Insecto , Hipertrofia , Iniciación de la Cadena Peptídica Traduccional/genética , Fenotipo , Unión Proteica , Interferencia de ARN , Transcriptoma/genética
11.
Science ; 350(6258): 339-42, 2015 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-26472911

RESUMEN

The Piwi-interacting RNA (piRNA) pathway is a small RNA-based innate immune system that defends germ cell genomes against transposons. In Drosophila ovaries, the nuclear Piwi protein is required for transcriptional silencing of transposons, though the precise mechanisms by which this occurs are unknown. Here we show that the CG9754 protein is a component of Piwi complexes that functions downstream of Piwi and its binding partner, Asterix, in transcriptional silencing. Enforced tethering of CG9754 to nascent messenger RNA transcripts causes cotranscriptional silencing of the source locus and the deposition of repressive chromatin marks. We have named CG9754 "Panoramix," and we propose that this protein could act as an adaptor, scaffolding interactions between the piRNA pathway and the general silencing machinery that it recruits to enforce transcriptional repression.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Silenciador del Gen , Proteínas Nucleares/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Proteínas Argonautas/metabolismo , Elementos Transponibles de ADN/genética , Proteínas de Drosophila/genética , Técnicas de Silenciamiento del Gen , Proteínas Nucleares/genética , Proteínas de Unión al ARN , Transcripción Genética
12.
Nat Cell Biol ; 17(5): 689-96, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25915123

RESUMEN

The differentiation of stem cells is a tightly regulated process essential for animal development and tissue homeostasis. Through this process, attainment of new identity and function is achieved by marked changes in cellular properties. Intrinsic cellular mechanisms governing stem cell differentiation remain largely unknown, in part because systematic forward genetic approaches to the problem have not been widely used. Analysing genes required for germline stem cell differentiation in the Drosophila ovary, we find that the mitochondrial ATP synthase plays a critical role in this process. Unexpectedly, the ATP synthesizing function of this complex was not necessary for differentiation, as knockdown of other members of the oxidative phosphorylation system did not disrupt the process. Instead, the ATP synthase acted to promote the maturation of mitochondrial cristae during differentiation through dimerization and specific upregulation of the ATP synthase complex. Taken together, our results suggest that ATP synthase-dependent crista maturation is a key developmental process required for differentiation independent of oxidative phosphorylation.


Asunto(s)
Diferenciación Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Mitocondrias/enzimología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Oocitos/enzimología , Fosforilación Oxidativa , Animales , Animales Modificados Genéticamente , Línea Celular , Bases de Datos Genéticas , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/ultraestructura , Genotipo , Microscopía Confocal , Microscopía Fluorescente , Mitocondrias/ultraestructura , ATPasas de Translocación de Protón Mitocondriales/genética , Oocitos/ultraestructura , Fenotipo , Multimerización de Proteína , Interferencia de ARN , Transducción de Señal , Transfección , Regulación hacia Arriba
13.
Mol Cell ; 50(5): 749-61, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23665227

RESUMEN

The Drosophila piRNA pathway provides an RNA-based immune system that defends the germline genome against selfish genetic elements. Two interrelated branches of the piRNA system exist: somatic cells that support oogenesis only employ Piwi, whereas germ cells utilize a more elaborate pathway centered on the three gonad-specific Argonaute proteins (Piwi, Aubergine, and Argonaute 3). While several key factors of each branch have been identified, our current knowledge is insufficient to explain the complex workings of the piRNA machinery. Here, we report a reverse genetic screen spanning the ovarian transcriptome in an attempt to uncover the full repertoire of genes required for piRNA-mediated transposon silencing in the female germline. Our screen reveals key factors of piRNA-mediated transposon silencing, including the piRNA biogenesis factors CG2183 (GASZ) and Deadlock. Our data uncover a previously unanticipated set of factors preferentially required for repression of different transposon types.


Asunto(s)
Elementos Transponibles de ADN , Drosophila melanogaster/genética , Perfilación de la Expresión Génica/métodos , Ovario/fisiología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Silenciador del Gen , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Factores de Iniciación de Péptidos/genética , Factores de Iniciación de Péptidos/metabolismo , ARN Interferente Pequeño/genética , Reproducibilidad de los Resultados
14.
RNA ; 18(8): 1446-57, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22753781

RESUMEN

In animals, the piRNA pathway preserves the integrity of gametic genomes, guarding them against the activity of mobile genetic elements. This innate immune mechanism relies on distinct genomic loci, termed piRNA clusters, to provide a molecular definition of transposons, enabling their discrimination from genes. piRNA clusters give rise to long, single-stranded precursors, which are processed into primary piRNAs through an unknown mechanism. These can engage in an adaptive amplification loop, the ping-pong cycle, to optimize the content of small RNA populations via the generation of secondary piRNAs. Many proteins have been ascribed functions in either primary biogenesis or the ping-pong cycle, though for the most part the molecular functions of proteins implicated in these pathways remain obscure. Here, we link shutdown (shu), a gene previously shown to be required for fertility in Drosophila, to the piRNA pathway. Analysis of knockdown phenotypes in both the germline and somatic compartments of the ovary demonstrate important roles for shutdown in both primary biogenesis and the ping-pong cycle. shutdown is a member of the FKBP family of immunophilins. Shu contains domains implicated in peptidyl-prolyl cis-trans isomerase activity and in the binding of HSP90-family chaperones, though the relevance of these domains to piRNA biogenesis is unknown.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Fertilidad/genética , Silenciador del Gen , Ovario/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Masculino , Ovario/citología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Retroelementos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
15.
Proc Natl Acad Sci U S A ; 108(8): 3204-9, 2011 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-21300896

RESUMEN

Small RNAs and a diverse array of protein partners control gene expression in eukaryotes through a variety of mechanisms. By combining siRNA affinity chromatography and mass spectrometry, we have identified the double-stranded RNA-binding domain protein Blanks to be an siRNA- and dsRNA-binding protein from Drosophila S2 cells. We find that Blanks is a nuclear factor that contributes to the efficiency of RNAi. Biochemical fractionation of a Blanks-containing complex shows that the Blanks complex is unlike previously described RNA-induced silencing complexes and associates with the DEAD-box helicase RM62, a protein previously implicated in RNA silencing. In flies, Blanks is highly expressed in testes tissues and is necessary for postmeiotic spermiogenesis, but loss of Blanks is not accompanied by detectable transposon derepression. Instead, genes related to innate immunity pathways are up-regulated in blanks mutant testes. These results reveal Blanks to be a unique component of a nuclear siRNA/dsRNA-binding complex that contributes to essential RNA silencing-related pathways in the male germ line.


Asunto(s)
Drosophila/fisiología , ARN Interferente Pequeño , Proteínas de Unión al ARN/fisiología , Espermatogénesis , Animales , Regulación de la Expresión Génica/fisiología , Inmunidad Innata/genética , Masculino , Interferencia de ARN , ARN Bicatenario , Testículo/química , Testículo/metabolismo
16.
Nat Cell Biol ; 11(9): 1150-6, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19684574

RESUMEN

Small RNAs direct RNA-induced silencing complexes (RISCs) to regulate stability and translation of mRNAs. RISCs associated with target mRNAs often accumulate in discrete cytoplasmic foci known as GW-bodies. However, RISC proteins can associate with membrane compartments such as the Golgi and endoplasmic reticulum. Here, we show that GW-bodies are associated with late endosomes (multivesicular bodies, MVBs). Blocking the maturation of MVBs into lysosomes by loss of the tethering factor HPS4 (ref. 5) enhances short interfering RNA (siRNA)- and micro RNA (miRNA)-mediated silencing in Drosophila melanogaster and humans. It also triggers over-accumulation of GW-bodies. Blocking MVB formation by ESCRT (endosomal sorting complex required for transport) depletion results in impaired miRNA silencing and loss of GW-bodies. These results indicate that active RISCs are physically and functionally coupled to MVBs. We further show that MVBs promote the competence of RISCs in loading small RNAs. We suggest that the recycling of RISCs is promoted by MVBs, resulting in RISCs more effectively engaging with small RNA effectors and possibly target RNAs. It may provide a means to enhance the dynamics of RNA silencing in the cytoplasm.


Asunto(s)
Drosophila melanogaster/metabolismo , Endosomas/metabolismo , Silenciador del Gen , ARN Interferente Pequeño/metabolismo , Animales , Transporte Biológico , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Células HeLa , Humanos , MicroARNs/metabolismo , Complejo Silenciador Inducido por ARN/metabolismo , Ubiquitinación
17.
Curr Biol ; 16(5): 530-5, 2006 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-16527750

RESUMEN

Short interfering RNAs (siRNAs) guide mRNA cleavage during RNA interference (RNAi). Only one siRNA strand assembles into the RNA-induced silencing complex (RISC), with preference given to the strand whose 5' terminus has lower base-pairing stability. In Drosophila, Dcr-2/R2D2 processes siRNAs from longer double-stranded RNAs (dsRNAs) and also nucleates RISC assembly, suggesting that nascent siRNAs could remain bound to Dcr-2/R2D2. In vitro, Dcr-2/R2D2 senses base-pairing asymmetry of synthetic siRNAs and dictates strand selection by asymmetric binding to the duplex ends. During dsRNA processing, Dicer (Dcr) liberates siRNAs from dsRNA ends in a manner dictated by asymmetric enzyme-substrate interactions. Because Dcr-2/R2D2 is unlikely to sense base-pairing asymmetry of an siRNA that is embedded within a precursor, it is not clear whether processed siRNAs strictly follow the thermodynamic asymmetry rules or whether processing polarity can affect strand selection. We use a Drosophila in vitro system in which defined siRNAs with known asymmetry can be generated from longer dsRNA precursors. These dsRNAs permit processing specifically from either the 5' or the 3' end of the thermodynamically favored strand of the incipient siRNA. Combined dsRNA-processing/mRNA-cleavage assays indicate that siRNA strand selection is independent of dsRNA processing polarity during Drosophila RISC assembly in vitro.


Asunto(s)
Drosophila/genética , Interferencia de ARN/fisiología , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Emparejamiento Base/genética , Proteínas de Drosophila/metabolismo , Modelos Genéticos , Óvulo/metabolismo , ARN Helicasas/metabolismo , ARN Mensajero/metabolismo , Complejo Silenciador Inducido por ARN/metabolismo , Ribonucleasa III , Termodinámica
18.
Cell ; 123(4): 543-5, 2005 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-16286001

RESUMEN

When an siRNA or miRNA proceeds through the RNA-induced silencing complex assembly pathway, only one of the two approximately 21-nucleotide RNA strands survives in the final, active complex. In this issue of Cell, Matranga et al. (2005) and Rand et al. (2005) reveal the fate of the rejected passenger siRNA strand. Additionally, Gregory et al. (2005) define a heterotrimeric complex from humans that appears to execute dsRNA loading, strand selection, and target mRNA cleavage activities.


Asunto(s)
Interferencia de ARN/fisiología , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/metabolismo , Complejo Silenciador Inducido por ARN/metabolismo , Animales , Proteínas Argonautas , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Modelos Genéticos , ARN Bicatenario/genética , ARN Interferente Pequeño/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA