Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nature ; 497(7449): 378-82, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23644459

ABSTRACT

MicroRNAs (miRNAs) show differential expression across breast cancer subtypes, and have both oncogenic and tumour-suppressive roles. Here we report the miRNA expression profiles of 1,302 breast tumours with matching detailed clinical annotation, long-term follow-up and genomic and messenger RNA expression data. This provides a comprehensive overview of the quantity, distribution and variation of the miRNA population and provides information on the extent to which genomic, transcriptional and post-transcriptional events contribute to miRNA expression architecture, suggesting an important role for post-transcriptional regulation. The key clinical parameters and cellular pathways related to the miRNA landscape are characterized, revealing context-dependent interactions, for example with regards to cell adhesion and Wnt signalling. Notably, only prognostic miRNA signatures derived from breast tumours devoid of somatic copy-number aberrations (CNA-devoid) are consistently prognostic across several other subtypes and can be validated in external cohorts. We then use a data-driven approach to seek the effects of miRNAs associated with differential co-expression of mRNAs, and find that miRNAs act as modulators of mRNA-mRNA interactions rather than as on-off molecular switches. We demonstrate such an important modulatory role for miRNAs in the biology of CNA-devoid breast cancers, a common subtype in which the immune response is prominent. These findings represent a new framework for studying the biology of miRNAs in human breast cancer.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Algorithms , Breast Neoplasms/pathology , DNA Copy Number Variations , Female , Follow-Up Studies , Gene Expression Profiling , Genome, Human/genetics , Humans , Kaplan-Meier Estimate , MicroRNAs/metabolism , Prognosis , Proportional Hazards Models , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
2.
Genome Res ; 24(1): 96-106, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24065776

ABSTRACT

Small RNA control of gene expression is critical for developmental processes in vertebrate embryos. To determine the dynamics of small RNA expression and to uncover novel small RNAs in the early vertebrate embryo, we performed high-throughput sequencing of all small RNAs in Xenopus tropicalis embryos at three developmental time points and in dissected halves of gastrula embryos. This analysis allowed us to identify novel microRNAs and we show that microRNA expression is highly dynamic and spatially localized in early embryos. In addition, we have developed a microRNA prediction pipeline and demonstrate that it has the power to predict new miRNAs that are experimentally detectable in frogs, mice, and humans. By combining the small RNA sequencing with mRNA profiling at the different developmental stages, we identify a new class of small noncoding RNAs that we name siteRNAs, which align in clusters to introns of protein-coding genes. We show that siteRNAs are derived from remnants of transposable elements present in the introns. We find that genes containing clusters of siteRNAs are transcriptionally repressed as compared with all genes. Furthermore, we show that this is true for individual genes containing siteRNA clusters, and that these genes are enriched in specific repressive histone modifications. Our data thus suggest a new mechanism of siteRNA-mediated gene silencing in vertebrates, and provide an example of how mobile elements can affect gene regulation.


Subject(s)
DNA Transposable Elements , Introns , MicroRNAs/genetics , MicroRNAs/metabolism , Xenopus Proteins/genetics , Xenopus/embryology , Xenopus/genetics , Animals , Embryo, Nonmammalian , Gastrula/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Silencing , High-Throughput Nucleotide Sequencing , Histones/metabolism , Humans , Mice , Sequence Analysis, RNA , Xenopus laevis/embryology , Xenopus laevis/genetics
3.
Genome Res ; 19(10): 1766-75, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19628731

ABSTRACT

Small regulatory RNAs have recently emerged as key regulators of eukaryotic gene expression. Here we used high-throughput sequencing to determine small RNA populations in the germline and soma of the African clawed frog Xenopus tropicalis. We identified a number of miRNAs that were expressed in the female germline. miRNA expression profiling revealed that miR-202-5p is an oocyte-enriched miRNA. We identified two novel miRNAs that were expressed in the soma. In addition, we sequenced large numbers of Piwi-associated RNAs (piRNAs) and other endogenous small RNAs, likely representing endogenous siRNAs (endo-siRNAs). Of these, only piRNAs were restricted to the germline, suggesting that endo-siRNAs are an abundant class of small RNAs in the vertebrate soma. In the germline, both endogenous small RNAs and piRNAs mapped to many high copy number loci. Furthermore, endogenous small RNAs mapped to the same specific subsets of repetitive elements in both the soma and the germline, suggesting that these RNAs might act to silence repetitive elements in both compartments. Data presented here suggest a conserved role for miRNAs in the vertebrate germline. Furthermore, this study provides a basis for the functional analysis of small regulatory RNAs in an important vertebrate model system.


Subject(s)
MicroRNAs/genetics , RNA, Small Interfering/genetics , RNA, Small Nuclear/genetics , Xenopus/genetics , Animals , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Germ Cells/metabolism , MicroRNAs/metabolism , RNA, Small Interfering/metabolism , RNA, Small Nuclear/metabolism , Signal Transduction/genetics , Vertebrates/genetics , Vertebrates/metabolism , Xenopus/metabolism
4.
Article in English | MEDLINE | ID: mdl-35338048

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the association between obesity, diabetes and metabolic related liver dysfunction and the incidence of cancer. DESIGN: This study was conducted with health record data available from the National Health Service in Tayside and Fife. Genetics of Diabetes Audit and Research Tayside, Scotland (GoDARTS), Scottish Health Research Register (SHARE) and Tayside and Fife diabetics, three Scottish cohorts of 13 695, 62 438 and 16 312 patients, respectively, were analysed in this study. Participants in GoDARTS were a volunteer sample, with half having type 2 diabetes mellitus(T2DM). SHARE was a volunteer sample. Tayside and Fife diabetics was a population-level cohort. Metabolic dysfunction-related liver disease (MDLD) was defined using alanine transaminase measurements, and individuals with alternative causes of liver disease (alcohol abuse, viruses, etc) were excluded from the analysis. RESULTS: MDLD associated with increased cancer incidence with a HR of 1.31 in a Cox proportional hazards model adjusted for sex, type 2 diabetes, body mass index(BMI), and smoking status (95% CI 1.27 to 1.35, p<0.0001). This was replicated in two further cohorts, and similar associations with cancer incidence were found for Fatty Liver Index (FLI), Fibrosis-4 Index (FIB-4) and non-alcoholic steatohepatitis (NASH). Homozygous carriers of the common non-alcoholic fatty liver disease (NAFLD) risk-variant PNPLA3 rs738409 had increased risk of cancer. (HR=1.27 (1.02 to 1.58), p=3.1×10 -2). BMI was not independently associated with cancer incidence when MDLD was included as a covariate. CONCLUSION: MDLD, FLI, FIB-4 and NASH associated with increased risk of cancer incidence and death. NAFLD may be a major component of the relationship between obesity and cancer incidence.


Subject(s)
Diabetes Mellitus, Type 2 , Metabolic Diseases , Neoplasms , Non-alcoholic Fatty Liver Disease , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Humans , Metabolic Diseases/complications , Neoplasms/complications , Neoplasms/etiology , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/epidemiology , Obesity/complications , Obesity/epidemiology , Risk Factors , State Medicine
5.
Biochemistry ; 50(35): 7514-21, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21815640

ABSTRACT

Several recent studies have provided evidence that LIN28, a cytoplasmic RNA-binding protein, inhibits the biogenesis of members of the let-7 microRNA family at the Dicer step in both mammals and Caenorhabditis elegans. However, the precise mechanism of inhibition is still poorly understood. Here we report on an in vitro study, which combined RNase footprinting, gel shift binding assays, and processing assays, to investigate the molecular basis and function of the interaction between the native let-7g precursor (pre-let-7g) and LIN28. We have mapped the structure of pre-let-7g and identified some regions of the terminal loop of pre-let-7g that physically interact with LIN28. We have also identified a conformational change upon LIN28 binding that results in the unwinding of an otherwise double-stranded region at the Dicer processing site of pre-let-7g. Furthermore, we showed that a mutant pre-let-7g that displays an open upper stem inhibited pre-let-7g Dicer processing to the same extent as LIN28. The data support a mechanism by which LIN28 can directly inhibit let-7g biogenesis at the Dicer processing step.


Subject(s)
MicroRNAs/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/physiology , Ribonuclease III/antagonists & inhibitors , Ribonuclease III/genetics , Animals , Base Sequence , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/genetics , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding/genetics , RNA Precursors/antagonists & inhibitors , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , Ribonuclease III/metabolism , Structure-Activity Relationship
6.
RNA ; 15(2): 337-45, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19144913

ABSTRACT

The Argonaute superfamily is a large family of RNA-binding proteins involved in gene regulation mediated by small noncoding RNA and characterized by the presence of PAZ and PIWI domains. The family consists of two branches, the Ago and the Piwi clade. Piwi proteins bind to 21-30-nucleotide-long Piwi-interacting RNAs (piRNAs), which map primarily to transposons and repeated sequence elements. Piwi/piRNAs are important regulators of gametogenesis and have been proposed to play roles in transposon silencing, DNA methylation, transcriptional silencing, and/or post-transcriptional control of translation and RNA stability. Most reports to date have concentrated on the Piwi family members in the male germline. We have identified four Piwi proteins in Xenopus and demonstrate that two, namely, Xiwi1b and Xili, are expressed in the oocyte and early embryo. Xiwi1 and Xili are predominantly found in small, separate complexes, and we do not detect significant interaction of Piwi proteins with the cap-binding complex. Putative nuclear localization and export signals were identified in Xiwi1 and Xili, supporting our observation that Xiwi1, but not Xili, is a nucleo-cytoplasmic protein. Furthermore, by immunoprecipitation of small RNAs, we establish Xiwi1 as a bona fide Piwi protein. These results suggest that the Piwi/piRNA pathway is active in translationally repressed oocytes. This is a significant finding as the Xenopus model provides an excellent tool to study post-transcriptional mechanisms.


Subject(s)
Oocytes/metabolism , Oogenesis , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus/physiology , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Embryonic Development , Female , Male , Molecular Sequence Data , Multiprotein Complexes/metabolism , Oocytes/growth & development , RNA-Binding Proteins/genetics , Xenopus/embryology , Xenopus/metabolism , Xenopus Proteins/genetics
7.
JAMA Cardiol ; 6(4): 379-386, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33326012

ABSTRACT

Importance: Sequencing studies have identified causal genetic variants for distinct subtypes of heart failure (HF) such as hypertrophic or dilated cardiomyopathy. However, the role of rare, high-impact variants in HF, for which ischemic heart disease is the leading cause, has not been systematically investigated. Objective: To assess the contribution of rare variants to all-cause HF with and without reduced left ventricular ejection fraction. Design, Setting, and Participants: This was a retrospective analysis of clinical trials and a prospective epidemiological resource (UK Biobank). Whole-exome sequencing of patients with HF was conducted from the Candesartan in Heart Failure-Assessment of Reduction in Mortality and Morbidity (CHARM) and Controlled Rosuvastatin Multinational Trial in Heart Failure (CORONA) clinical trials. Data were collected from March 1999 to May 2003 for the CHARM studies and September 2003 to July 2007 for the CORONA study. Using a gene-based collapsing approach, the proportion of patients with HF and controls carrying rare and presumed deleterious variants was compared. The burden of pathogenic variants in known cardiomyopathy genes was also investigated to assess the diagnostic yield. Exome sequencing data were generated between January 2018 and October 2018, and analysis began October 2018 and ended April 2020. Main Outcomes and Measures: Odds ratios and P values for genes enriched for rare and presumed deleterious variants in either patients with HF or controls and diagnostic yield of pathogenic variants in known cardiomyopathy genes. Results: This study included 5942 patients with HF and 13 156 controls. The mean (SD) age was 68.9 (9.9) years and 4213 (70.9%) were male. A significant enrichment of protein-truncating variants in the TTN gene (P = 3.35 × 10-13; odds ratio, 2.54; 95% CI, 1.96-3.31) that was further increased after restriction to variants in exons constitutively expressed in the heart (odds ratio, 4.52; 95% CI, 3.10-6.68). Validation using UK Biobank data showed a similar enrichment (odds ratio, 4.97; 95% CI, 3.94-6.19 after restriction). In the clinical trials, 201 of 5916 patients with HF (3.4%) had a pathogenic or likely pathogenic cardiomyopathy variant implicating 21 different genes. Notably, 121 of 201 individuals (60.2%) had ischemic heart disease as the clinically identified etiology for the HF. Individuals with HF and preserved ejection fraction had only a slightly lower yield than individuals with midrange or reduced ejection fraction (20 of 767 [2.6%] vs 15 of 392 [3.8%] vs 166 of 4757 [3.5%]). Conclusions and Relevance: An increased burden of diagnostic mendelian cardiomyopathy variants in a broad group of patients with HF of mostly ischemic etiology compared with controls was observed. This work provides further evidence that mendelian genetic conditions may represent an important subset of complex late-onset diseases such as HF, irrespective of the clinical presentation.


Subject(s)
Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Heart Failure/genetics , Aged , Case-Control Studies , Female , Humans , Male , Retrospective Studies , Exome Sequencing
8.
Circ Genom Precis Med ; 13(6): e003030, 2020 12.
Article in English | MEDLINE | ID: mdl-33125268

ABSTRACT

BACKGROUND: Spontaneous coronary artery dissection (SCAD) occurs when an epicardial coronary artery is narrowed or occluded by an intramural hematoma. SCAD mainly affects women and is associated with pregnancy and systemic arteriopathies, particularly fibromuscular dysplasia. Variants in several genes, such as those causing connective tissue disorders, have been implicated; however, the genetic architecture is poorly understood. Here, we aim to better understand the diagnostic yield of rare variant genetic testing among a cohort of SCAD survivors and to identify genes or gene sets that have a significant enrichment of rare variants. METHODS: We sequenced a cohort of 384 SCAD survivors from the United Kingdom, alongside 13 722 UK Biobank controls and a validation cohort of 92 SCAD survivors. We performed a research diagnostic screen for pathogenic variants and exome-wide and gene-set rare variant collapsing analyses. RESULTS: The majority of patients within both cohorts are female, 29% of the study cohort and 14% validation cohort have a remote arteriopathy. Four cases across the 2 cohorts had a diagnosed connective tissue disorder. We identified pathogenic or likely pathogenic variants in 7 genes (PKD1, COL3A1, SMAD3, TGFB2, LOX, MYLK, and YY1AP1) in 14/384 cases in the study cohort and in 1/92 cases in the validation cohort. In our rare variant collapsing analysis, PKD1 was the highest-ranked gene, and several functionally plausible genes were enriched for rare variants, although no gene achieved study-wide statistical significance. Gene-set enrichment analysis suggested a role for additional genes involved in renal function. CONCLUSIONS: By studying the largest sequenced cohort of SCAD survivors, we demonstrate that, based on current knowledge, only a small proportion have a pathogenic variant that could explain their disease. Our findings strengthen the overlap between SCAD and renal and connective tissue disorders, and we highlight several new genes for future validation.


Subject(s)
Coronary Vessel Anomalies/genetics , Exome Sequencing , Genetic Variation , Genome, Human , Vascular Diseases/congenital , Adult , Aged , Cohort Studies , Female , Humans , Machine Learning , Male , Middle Aged , Models, Genetic , United Kingdom , Vascular Diseases/genetics , Young Adult
9.
PLoS One ; 12(5): e0175779, 2017.
Article in English | MEDLINE | ID: mdl-28472036

ABSTRACT

Personalized healthcare relies on accurate companion diagnostic assays that enable the most appropriate treatment decision for cancer patients. Extensive assay validation prior to use in a clinical setting is essential for providing a reliable test result. This poses a challenge for low prevalence mutations with limited availability of appropriate clinical samples harboring the mutation. To enable prospective screening for the low prevalence AKT1 E17K mutation, we have developed and validated a competitive allele-specific TaqMan® PCR (castPCR™) assay for mutation detection in formalin-fixed paraffin-embedded (FFPE) tumor tissue. Analysis parameters of the castPCR™ assay were established using an FFPE DNA reference standard and its analytical performance was assessed using 338 breast cancer and gynecological cancer FFPE samples. With recent technical advances for minimally invasive mutation detection in circulating tumor DNA (ctDNA), we subsequently also evaluated the OncoBEAM™ assay to enable plasma specimens as additional diagnostic opportunity for AKT1 E17K mutation testing. The analysis performance of the OncoBEAM™ test was evaluated using a novel AKT1 E17K ctDNA reference standard consisting of sheared genomic DNA spiked into human plasma. Both assays are employed at centralized testing laboratories operating according to quality standards for prospective identification of the AKT1 E17K mutation in ER+ breast cancer patients in the context of a clinical trial evaluating the AKT inhibitor AZD5363 in combination with endocrine (fulvestrant) therapy.


Subject(s)
Mutation , Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/genetics , Humans , Neoplasms/blood , Neoplasms/genetics , Paraffin Embedding , Polymerase Chain Reaction , Reference Standards , Reproducibility of Results
10.
Nucleic Acids Res ; 32(3): 1242-50, 2004.
Article in English | MEDLINE | ID: mdl-14973223

ABSTRACT

Mer1p activates the splicing of at least three pre-mRNAs (AMA1, MER2, MER3) during meiosis in the yeast Saccharomyces cerevisiae. We demonstrate that enhancer recognition by Mer1p is separable from Mer1p splicing activation. The C-terminal KH-type RNA-binding domain of Mer1p recognizes introns that contain the Mer1p splicing enhancer, while the N-terminal domain interacts with the spliceosome and activates splicing. Prior studies have implicated the U1 snRNP and recognition of the 5' splice site as key elements in Mer1p-activated splicing. We provide new evidence that Mer1p may also function at later steps of spliceosome assembly. First, Mer1p can activate splicing of introns that have mutated branch point sequences. Secondly, Mer1p fails to activate splicing in the absence of the non-essential U2 snRNP protein Snu17p. Thirdly, Mer1p interacts with the branch point binding proteins Mud2p and Bbp1p and the U2 snRNP protein Prp11p by two-hybrid assays. We conclude that Mer1p is a modular splicing regulator that can activate splicing at several early steps of spliceosome assembly and depends on the activities of both U1 and U2 snRNP proteins to activate splicing.


Subject(s)
RNA Splicing , RNA-Binding Proteins/physiology , Ribonucleoprotein, U2 Small Nuclear/physiology , Saccharomyces cerevisiae Proteins/physiology , Base Sequence , Introns , Mutation , Protein Structure, Tertiary , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , Ribonucleoprotein, U1 Small Nuclear/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Spliceosomes/metabolism
11.
Curr Protoc Mol Biol ; 112: 7.21.1-7.21.23, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26423591

ABSTRACT

Target enrichment is commonly used in next generation sequencing (NGS) workflows to eliminate genomic DNA regions that are not of interest for a particular experiment. By only targeting specific regions such as exons, one can obtain greater depth of DNA sequencing coverage for regions of interest or increase the sampling numbers of individuals, thereby saving both time and cost. This overview of target enrichment strategies provides a high-level review of distinct approaches to capture specific sequences: (a) hybridization-based strategies, (b) transposon-mediated fragmentation (tagmentation), (c) molecular inversion probes (MIPs), and (d) singleplex and multiplex polymerase chain reaction (PCR) target enrichment. Strategies for assay design and performance criteria are also discussed. Other platforms currently in development are also briefly described.


Subject(s)
Gene Targeting/methods , Nucleic Acid Amplification Techniques/methods , High-Throughput Nucleotide Sequencing/methods
12.
Stem Cells Dev ; 21(2): 181-90, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-21504359

ABSTRACT

Amphibian oocytes have the special ability to directly induce the transcription of pluripotency and other genes in transplanted somatic nuclei. To this extent, oocytes induce a stem cell-like pattern of transcription in somatic cell nuclei. We ask whether the induced transcription in transplanted nuclei reflects the normal transcriptional activity of oocyte genes. We describe here the transcript content of a wide range of genes in Xenopus tropicalis oocytes. Using accurate quantitation, we find that each mature oocyte has accumulated several hundred transcripts of cell-type specific genes. This value is several orders of magnitude greater than the "leakage" level found in most somatic cells and about the same level found in somatic cells where these genes are fully expressed. Illumina sequencing confirms the high transcript content of a mature Xenopus oocyte. Most of the transcripts from these highly expressed genes in oocytes are correctly and efficiently spliced. Our results contribute a more quantitative view of certain amphibian oocyte transcripts than previously available. Our results also show that transplanted somatic nuclei conform, with respect to the genes analyzed, to the transcriptional characteristics of the recipient oocytes.


Subject(s)
Cell Nucleus/genetics , Nuclear Transfer Techniques , Oocytes , RNA, Messenger/genetics , Transcriptome/genetics , Xenopus Proteins/genetics , Animals , Cell Nucleus/metabolism , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Oocytes/cytology , Oocytes/metabolism , RNA Splicing , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcriptional Activation , Xenopus , Xenopus Proteins/metabolism
13.
PLoS One ; 7(3): e33750, 2012.
Article in English | MEDLINE | ID: mdl-22448270

ABSTRACT

The genes alg-1 and alg-2 (referred to as "alg-1/2") encode the Argonaute proteins affiliated to the microRNA (miRNA) pathway in C. elegans. Bound to miRNAs they form the effector complex that effects post-transcriptional gene silencing. In order to define biological features important to understand the mode of action of these Argonautes, we characterize aspects of these genes during development. We establish that alg-1/2 display an overlapping spatio-temporal expression profile and shared association to a miRNAs set, but with gene-specific predominant expression in various cells and increased relative association to defined miRNAs. Congruent with their spatio-temporal coincidence and regardless of alg-1/2 drastic post-embryonic differences, only loss of both genes leads to embryonic lethality. Embryos without zygotic alg-1/2 predominantly arrest during the morphogenetic process of elongation with defects in the epidermal-muscle attachment structures. Altogether our results highlight similarities and specificities of the alg-1/2 likely to be explained at different cellular and molecular levels.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Epidermal Cells , Epidermis/metabolism , Molecular Sequence Data , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , RNA-Binding Proteins/metabolism , Sequence Homology, Amino Acid
14.
Nat Neurosci ; 15(1): 29-38, 2011 Dec 04.
Article in English | MEDLINE | ID: mdl-22138647

ABSTRACT

During axon pathfinding, growth cones commonly show changes in sensitivity to guidance cues that follow a cell-intrinsic timetable. The cellular timer mechanisms that regulate such changes are, however, poorly understood. Here we have investigated microRNAs (miRNAs) in the timing control of sensitivity to the semaphorin Sema3A in Xenopus laevis retinal ganglion cell (RGC) growth cones. A developmental profiling screen identified miR-124 as a candidate timer. Loss of miR-124 delayed the onset of Sema3A sensitivity and concomitant neuropilin-1 (NRP1) receptor expression and caused cell-autonomous pathfinding errors. CoREST, a cofactor of a NRP1 repressor, was newly identified as a target and mediator of miR-124 for this highly specific temporal aspect of RGC growth cone responsiveness. Our findings indicate that miR-124 is important in regulating the intrinsic temporal changes in RGC growth cone sensitivity and suggest that miRNAs may act broadly as linear timers in vertebrate neuronal development.


Subject(s)
Cell Movement/physiology , Growth Cones/metabolism , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Retinal Ganglion Cells/metabolism , Semaphorin-3A/metabolism , Animals , Axons/metabolism , Cell Differentiation/physiology , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Neuropilin-1/genetics , Neuropilin-1/metabolism , Repressor Proteins/genetics , Semaphorin-3A/genetics , Signal Transduction/physiology , Xenopus laevis
15.
Genetics ; 185(3): 897-905, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20421599

ABSTRACT

microRNAs (miRNAs) are approximately 22-nucleotide small RNAs that act as endogenous regulators of gene expression by base-pairing with target mRNAs. Here we analyze the function of the six members of the Caenorhabditis elegans miR-51 family of miRNAs (miR-51, miR-52, miR-53, miR-54, miR-55, miR-56). miR-51 family miRNAs are broadly expressed from mid-embryogenesis onward. The miR-51 family is redundantly required for embryonic development. mir-51 family mutants display a highly penetrant pharynx unattached (Pun) phenotype, where the pharyngeal muscle, the food pump of C. elegans, is not attached to the mouth. Unusually, the Pun phenotype in mir-51 family mutants is not due to a failure to attach, but instead a failure to maintain attachment during late embryogenesis. Expression of the miR-51 family in the mouth is sufficient to maintain attachment. The Fat cadherin ortholog CDH-3 is expressed in the mouth and is a direct target of the miR-51 family miRNAs. Genetic analysis reveals that miR-51 family miRNAs might act in part through CDH-3 to regulate pharynx attachment. This study is the first to assign a function to the miR-51/miR-100 miRNA family in any organism.


Subject(s)
Caenorhabditis elegans/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Pharynx/metabolism , Animals , Base Sequence , Cadherins/genetics , Cadherins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Dermis/cytology , Dermis/metabolism , Fluorescent Antibody Technique , Molecular Sequence Data , Morphogenesis/physiology , Mutation/genetics , Pharynx/embryology , Phenotype , Sequence Homology, Nucleic Acid
16.
Nat Struct Mol Biol ; 16(10): 1016-20, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19713957

ABSTRACT

The let-7 microRNA (miRNA) is an ultraconserved regulator of stem cell differentiation and developmental timing and a candidate tumor suppressor. Here we show that LIN-28 and the poly(U) polymerase PUP-2 regulate let-7 processing in Caenorhabditis elegans. We demonstrate that lin-28 is necessary and sufficient to block let-7 activity in vivo; LIN-28 directly binds let-7 pre-miRNA to prevent Dicer processing. Moreover, we have identified a poly(U) polymerase, PUP-2, which regulates the stability of LIN-28-blockaded let-7 pre-miRNA and contributes to LIN-28-dependent regulation of let-7 during development. We show that PUP-2 and LIN-28 interact directly, and that LIN-28 stimulates uridylation of let-7 pre-miRNA by PUP-2 in vitro. Our results demonstrate that LIN-28 and let-7 form an ancient regulatory switch, conserved from nematodes to humans, and provide insight into the mechanism of LIN-28 action in vivo. Uridylation by a PUP-2 ortholog might regulate let-7 and additional miRNAs in other species. Given the roles of Lin28 and let-7 in stem cell and cancer biology, we propose that such poly(U) polymerases are potential therapeutic targets.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Gene Expression Regulation , MicroRNAs/genetics , Nucleotidyltransferases/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Animals, Genetically Modified , Base Sequence , Caenorhabditis elegans , Cell Differentiation , MicroRNAs/metabolism , Models, Biological , Molecular Sequence Data , Nucleotidyltransferases/metabolism , RNA Processing, Post-Transcriptional , Ribonuclease III/metabolism , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL