Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Am J Hum Genet ; 111(6): 1006-1017, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38703768

ABSTRACT

We present shaPRS, a method that leverages widespread pleiotropy between traits or shared genetic effects across ancestries, to improve the accuracy of polygenic scores. The method uses genome-wide summary statistics from two diseases or ancestries to improve the genetic effect estimate and standard error at SNPs where there is homogeneity of effect between the two datasets. When there is significant evidence of heterogeneity, the genetic effect from the disease or population closest to the target population is maintained. We show via simulation and a series of real-world examples that shaPRS substantially enhances the accuracy of polygenic risk scores (PRSs) for complex diseases and greatly improves PRS performance across ancestries. shaPRS is a PRS pre-processing method that is agnostic to the actual PRS generation method, and as a result, it can be integrated into existing PRS generation pipelines and continue to be applied as more performant PRS methods are developed over time.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Multifactorial Inheritance , Polymorphism, Single Nucleotide , Multifactorial Inheritance/genetics , Humans , Models, Genetic , Computer Simulation , Genetic Pleiotropy , Phenotype
2.
Bioinformatics ; 39(7)2023 07 01.
Article in English | MEDLINE | ID: mdl-37338536

ABSTRACT

MOTIVATION: While many pipelines have been developed for calling genotypes using RNA-sequencing (RNA-Seq) data, they all have adapted DNA genotype callers that do not model biases specific to RNA-Seq such as allele-specific expression (ASE). RESULTS: Here, we present Bayesian beta-binomial mixture model (BBmix), a Bayesian beta-binomial mixture model that first learns the expected distribution of read counts for each genotype, and then deploys those learned parameters to call genotypes probabilistically. We benchmarked our model on a wide variety of datasets and showed that our method generally performed better than competitors, mainly due to an increase of up to 1.4% in the accuracy of heterozygous calls, which may have a big impact in reducing false positive rate in applications sensitive to genotyping error such as ASE. Moreover, BBmix can be easily incorporated into standard pipelines for calling genotypes. We further show that parameters are generally transferable within datasets, such that a single learning run of less than 1 h is sufficient to call genotypes in a large number of samples. AVAILABILITY AND IMPLEMENTATION: We implemented BBmix as an R package that is available for free under a GPL-2 licence at https://gitlab.com/evigorito/bbmix and https://cran.r-project.org/package=bbmix with accompanying pipeline at https://gitlab.com/evigorito/bbmix_pipeline.


Subject(s)
High-Throughput Nucleotide Sequencing , RNA , Genotype , Bayes Theorem , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods , RNA/genetics , Software
3.
Bioinformatics ; 37(23): 4444-4450, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34145897

ABSTRACT

MOTIVATION: Polygenic scores (PGS) aim to genetically predict complex traits at an individual level. PGS are typically trained on genome-wide association summary statistics and require an independent test dataset to tune parameters. More recent methods allow parameters to be tuned on the training data, removing the need for independent test data, but approaches are computationally intensive. Based on fine-mapping principles, we present RápidoPGS, a flexible and fast method to compute PGS requiring summary-level Genome-wide association studies (GWAS) datasets only, with little computational requirements and no test data required for parameter tuning. RESULTS: We show that RápidoPGS performs slightly less well than two out of three other widely used PGS methods (LDpred2, PRScs and SBayesR) for case-control datasets, with median r2 difference: -0.0092, -0.0042 and 0.0064, respectively, but up to 17 000-fold faster with reduced computational requirements. RápidoPGS is implemented in R and can work with user-supplied summary statistics or download them from the GWAS catalog. AVAILABILITY AND IMPLEMENTATION: Our method is available with a GPL license as an R package from CRAN and GitHub. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Genome-Wide Association Study , Multifactorial Inheritance , Genotype , Polymorphism, Single Nucleotide
4.
Am J Hum Genet ; 108(6): 983-1000, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33909991

ABSTRACT

We present EPISPOT, a fully joint framework which exploits large panels of epigenetic annotations as variant-level information to enhance molecular quantitative trait locus (QTL) mapping. Thanks to a purpose-built Bayesian inferential algorithm, EPISPOT accommodates functional information for both cis and trans actions, including QTL hotspot effects. It effectively couples simultaneous QTL analysis of thousands of genetic variants and molecular traits with hypothesis-free selection of biologically interpretable annotations which directly contribute to the QTL effects. This unified, epigenome-aided learning boosts statistical power and sheds light on the regulatory basis of the uncovered hits; EPISPOT therefore marks an essential step toward improving the challenging detection and functional interpretation of trans-acting genetic variants and hotspots. We illustrate the advantages of EPISPOT in simulations emulating real-data conditions and in a monocyte expression QTL study, which confirms known hotspots and finds other signals, as well as plausible mechanisms of action. In particular, by highlighting the role of monocyte DNase-I sensitivity sites from >150 epigenetic annotations, we clarify the mediation effects and cell-type specificity of major hotspots close to the lysozyme gene. Our approach forgoes the daunting and underpowered task of one-annotation-at-a-time enrichment analyses for prioritizing cis and trans QTL hits and is tailored to any transcriptomic, proteomic, or metabolomic QTL problem. By enabling principled epigenome-driven QTL mapping transcriptome-wide, EPISPOT helps progress toward a better functional understanding of genetic regulation.


Subject(s)
Algorithms , Computer Simulation , Epigenome , Models, Genetic , Mutation , Phenotype , Quantitative Trait Loci , Bayes Theorem , Chromosome Mapping , Humans
5.
Nat Comput Sci ; 1: 421-432, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34993494

ABSTRACT

Detecting genetic variants associated with traits (quantitative trait loci, QTL) requires genotyped study individuals. Here we describe BaseQTL, a Bayesian method that exploits allele-specific expression to map molecular QTL from sequencing reads (eQTL for gene expression) even when no genotypes are available. When used with genotypes to map eQTL, BaseQTL has lower error rates and increased power compared with existing QTL mapping methods. Running without genotypes limits how many tests can be performed, but due to the proximity of QTL variants to gene bodies, the 2.8% of variants within a 100 kB window that could be tested contained 26% of eQTL detectable with genotypes. eQTL effect estimates were invariably consistent between analyses performed with and without genotypes. Often, sequencing data may be generated in the absence of genotypes on patients and controls in differential expression studies, and we identified an apparent psoriasis-specific eQTL for GSTP1 in one such dataset, providing new insights into disease-dependent gene regulation.

6.
Life Sci Alliance ; 2(3)2019 06.
Article in English | MEDLINE | ID: mdl-31097471

ABSTRACT

A fast antibody response can be critical to contain rapidly dividing pathogens. This can be achieved by the expansion of antigen-specific B cells in response to T-cell help followed by differentiation into plasmablasts. MicroRNA-155 (miR-155) is required for optimal T-cell-dependent extrafollicular responses via regulation of PU.1, although the cellular processes underlying this defect are largely unknown. Here, we show that miR-155 regulates the early expansion of B-blasts and later on the survival and proliferation of plasmablasts in a B-cell-intrinsic manner, by tracking antigen-specific B cells in vivo since the onset of antigen stimulation. In agreement, comparative analysis of the transcriptome of miR-155-sufficient and miR-155-deficient plasmablasts at the peak of the response showed that the main processes regulated by miR-155 were DNA metabolic process, DNA replication, and cell cycle. Thus, miR-155 controls the extent of the extrafollicular response by regulating the survival and proliferation of B-blasts, plasmablasts and, consequently, antibody production.


Subject(s)
B-Lymphocyte Subsets/metabolism , MicroRNAs/genetics , Plasma Cells/metabolism , Animals , Antibody Formation/genetics , Antibody Formation/immunology , B-Lymphocyte Subsets/immunology , Biomarkers , Cell Proliferation , Cell Survival/genetics , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Plasma Cells/immunology
7.
PLoS One ; 11(7): e0158801, 2016.
Article in English | MEDLINE | ID: mdl-27463617

ABSTRACT

Population-based genome wide association studies have identified a locus at 9p22.2 associated with ovarian cancer risk, which also modifies ovarian cancer risk in BRCA1 and BRCA2 mutation carriers. We conducted fine-scale mapping at 9p22.2 to identify potential causal variants in BRCA1 and BRCA2 mutation carriers. Genotype data were available for 15,252 (2,462 ovarian cancer cases) BRCA1 and 8,211 (631 ovarian cancer cases) BRCA2 mutation carriers. Following genotype imputation, ovarian cancer associations were assessed for 4,873 and 5,020 SNPs in BRCA1 and BRCA 2 mutation carriers respectively, within a retrospective cohort analytical framework. In BRCA1 mutation carriers one set of eight correlated candidate causal variants for ovarian cancer risk modification was identified (top SNP rs10124837, HR: 0.73, 95%CI: 0.68 to 0.79, p-value 2× 10-16). These variants were located up to 20 kb upstream of BNC2. In BRCA2 mutation carriers one region, up to 45 kb upstream of BNC2, and containing 100 correlated SNPs was identified as candidate causal (top SNP rs62543585, HR: 0.69, 95%CI: 0.59 to 0.80, p-value 1.0 × 10-6). The candidate causal in BRCA1 mutation carriers did not include the strongest associated variant at this locus in the general population. In sum, we identified a set of candidate causal variants in a region that encompasses the BNC2 transcription start site. The ovarian cancer association at 9p22.2 may be mediated by different variants in BRCA1 mutation carriers and in the general population. Thus, potentially different mechanisms may underlie ovarian cancer risk for mutation carriers and the general population.


Subject(s)
Chromosomes, Human, Pair 9 , Genes, BRCA1 , Genes, BRCA2 , Genetic Carrier Screening , Genetic Predisposition to Disease , Ovarian Neoplasms/genetics , Chromosome Mapping , Female , Humans , Polymorphism, Single Nucleotide
8.
J Clin Invest ; 126(1): 377-88, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26657861

ABSTRACT

The production of high-affinity antibodies by B cells is essential for pathogen clearance. Antibody affinity for antigen is increased through the affinity maturation in germinal centers (GCs). This is an iterative process in which B cells cycle between proliferation coupled with the acquisition of mutations and antigen-based positive selection, resulting in retention of the highest-affinity B cell clones. The posttranscriptional regulator microRNA-155 (miR-155) is critical for efficient affinity maturation and the maintenance of the GCs; however, the cellular and molecular mechanism by which miR-155 regulates GC responses is not well understood. Here, we utilized a miR-155 reporter mouse strain and showed that miR-155 is coexpressed with the proto-oncogene encoding c-MYC in positively selected B cells. Functionally, miR-155 protected positively selected c-MYC+ B cells from apoptosis, allowing clonal expansion of this population, providing an explanation as to why Mir155 deletion impairs affinity maturation and promotes the premature collapse of GCs. We determined that miR-155 directly inhibits the Jumonji family member JARID2, which enhances B cell apoptosis when overexpressed, and thereby promotes GC B cell survival. Our findings also suggest that there is cooperation between c-MYC and miR-155 during the normal GC response, a cooperation that may explain how c-MYC and miR-155 can collaboratively function as oncogenes.


Subject(s)
Apoptosis , B-Lymphocytes/physiology , MicroRNAs/physiology , Proto-Oncogene Proteins c-myc/analysis , Animals , Cell Survival , Germinal Center/physiology , Mice , Mice, Inbred C57BL , MicroRNAs/analysis , Polycomb Repressive Complex 2/physiology
9.
Proc Natl Acad Sci U S A ; 112(23): 7231-6, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-25995365

ABSTRACT

The response to an innate immune challenge is conditioned by the time of day, but the molecular basis for this remains unclear. In myeloid cells, there is a temporal regulation to induction by lipopolysaccharide (LPS) of the proinflammatory microRNA miR-155 that correlates inversely with levels of BMAL1. BMAL1 in the myeloid lineage inhibits activation of NF-κB and miR-155 induction and protects mice from LPS-induced sepsis. Bmal1 has two miR-155-binding sites in its 3'-UTR, and, in response to LPS, miR-155 binds to these two target sites, leading to suppression of Bmal1 mRNA and protein in mice and humans. miR-155 deletion perturbs circadian function, gives rise to a shorter circadian day, and ablates the circadian effect on cytokine responses to LPS. Thus, the molecular clock controls miR-155 induction that can repress BMAL1 directly. This leads to an innate immune response that is variably responsive to challenges across the circadian day.


Subject(s)
ARNTL Transcription Factors/physiology , Circadian Rhythm , Immunity, Innate , Macrophages/immunology , MicroRNAs/physiology , 3' Untranslated Regions , ARNTL Transcription Factors/genetics , Adipose Tissue/metabolism , Animals , Cytokines/biosynthesis , Macrophages/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism
10.
J Exp Med ; 211(11): 2183-98, 2014 Oct 20.
Article in English | MEDLINE | ID: mdl-25288398

ABSTRACT

A single microRNA (miRNA) can regulate the expression of many genes, though the level of repression imparted on any given target is generally low. How then is the selective pressure for a single miRNA/target interaction maintained across long evolutionary distances? We addressed this problem by disrupting in vivo the interaction between miR-155 and PU.1 in mice. Remarkably, this interaction proved to be key to promoting optimal T cell-dependent B cell responses, a previously unrecognized role for PU.1. Mechanistically, miR-155 inhibits PU.1 expression, leading to Pax5 down-regulation and the initiation of the plasma cell differentiation pathway. Additional PU.1 targets include a network of genes whose products are involved in adhesion, with direct links to B-T cell interactions. We conclude that the evolutionary adaptive selection of the miR-155-PU.1 interaction is exercised through the effectiveness of terminal B cell differentiation.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , MicroRNAs/genetics , PAX5 Transcription Factor/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , 3' Untranslated Regions , Animals , Antibody Formation/genetics , Antibody Formation/immunology , B-Lymphocytes/immunology , Base Sequence , Binding Sites , Cell Adhesion/genetics , Cell Communication/genetics , Cell Communication/immunology , Gene Expression Regulation , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphopoiesis/genetics , Mice , Mice, Knockout , MicroRNAs/chemistry , Myelopoiesis/genetics , PAX5 Transcription Factor/chemistry , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Trans-Activators/chemistry , Transcription Factors/genetics
11.
Nat Immunol ; 15(6): 484-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24840979

ABSTRACT

The rapid changes in gene expression that accompany developmental transitions, stress responses and proliferation are controlled by signal-mediated coordination of transcriptional and post-transcriptional mechanisms. In recent years, understanding of the mechanics of these processes and the contexts in which they are employed during hematopoiesis and immune challenge has increased. An important aspect of this progress is recognition of the importance of RNA-binding proteins and noncoding RNAs. These have roles in the development and function of the immune system and in pathogen life cycles, and they represent an important aspect of intracellular immunity.


Subject(s)
Immune System/immunology , Immunity, Cellular/genetics , Lymphocyte Activation/genetics , RNA, Long Noncoding/genetics , RNA, Small Untranslated/genetics , Animals , Gene Expression/immunology , Hematopoiesis/genetics , Humans , Immunity, Cellular/immunology , Lymphocyte Activation/immunology , Mice , RNA Processing, Post-Transcriptional/genetics , RNA Processing, Post-Transcriptional/immunology , RNA-Binding Proteins/genetics , Signal Transduction/genetics , Transcription, Genetic
12.
J Immunol ; 191(6): 3037-3048, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23960236

ABSTRACT

MicroRNAs (MiRs) are small, noncoding RNAs that regulate gene expression posttranscriptionally. In this study, we show that MiR-210 is induced by Oct-2, a key transcriptional mediator of B cell activation. Germline deletion of MiR-210 results in the development of autoantibodies from 5 mo of age. Overexpression of MiR-210 in vivo resulted in cell autonomous expansion of the B1 lineage and impaired fitness of B2 cells. Mice overexpressing MiR-210 exhibited impaired class-switched Ab responses, a finding confirmed in wild-type B cells transfected with a MiR-210 mimic. In vitro studies demonstrated defects in cellular proliferation and cell cycle entry, which were consistent with the transcriptomic analysis demonstrating downregulation of genes involved in cellular proliferation and B cell activation. These findings indicate that Oct-2 induction of MiR-210 provides a novel inhibitory mechanism for the control of B cells and autoantibody production.


Subject(s)
Autoantibodies/biosynthesis , B-Lymphocytes/metabolism , Lymphocyte Activation/immunology , MicroRNAs/biosynthesis , Octamer Transcription Factor-2/metabolism , Animals , Autoantibodies/immunology , B-Lymphocytes/immunology , Cell Separation , Chromatin Immunoprecipitation , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/immunology , Octamer Transcription Factor-2/immunology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Transcriptome
13.
Circulation ; 128(13): 1420-32, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23956210

ABSTRACT

BACKGROUND: Cardiac hypertrophy and subsequent heart failure triggered by chronic hypertension represent major challenges for cardiovascular research. Beyond neurohormonal and myocyte signaling pathways, growing evidence suggests inflammatory signaling pathways as therapeutically targetable contributors to this process. We recently reported that microRNA-155 is a key mediator of cardiac inflammation and injury in infectious myocarditis. Here, we investigated the impact of microRNA-155 manipulation in hypertensive heart disease. METHODS AND RESULTS: Genetic loss or pharmacological inhibition of the leukocyte-expressed microRNA-155 in mice markedly reduced cardiac inflammation, hypertrophy, and dysfunction on pressure overload. These alterations were macrophage dependent because in vivo cardiomyocyte-specific microRNA-155 manipulation did not affect cardiac hypertrophy or dysfunction, whereas bone marrow transplantation from wild-type mice into microRNA-155 knockout animals rescued the hypertrophic response of the cardiomyocytes and vice versa. In vitro, media from microRNA-155 knockout macrophages blocked the hypertrophic growth of stimulated cardiomyocytes, confirming that macrophages influence myocyte growth in a microRNA-155-dependent paracrine manner. These effects were at least partly mediated by the direct microRNA-155 target suppressor of cytokine signaling 1 (Socs1) because Socs1 knockdown in microRNA-155 knockout macrophages largely restored their hypertrophy-stimulating potency. CONCLUSIONS: Our findings reveal that microRNA-155 expression in macrophages promotes cardiac inflammation, hypertrophy, and failure in response to pressure overload. These data support the causative significance of inflammatory signaling in hypertrophic heart disease and demonstrate the feasibility of therapeutic microRNA targeting of inflammation in heart failure.


Subject(s)
Cardiomegaly/pathology , Heart Failure/pathology , Macrophages/pathology , MicroRNAs/genetics , Myocytes, Cardiac/pathology , Animals , Cardiomegaly/genetics , Cells, Cultured , Heart Failure/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Rats
14.
Immunol Rev ; 253(1): 146-57, 2013 May.
Article in English | MEDLINE | ID: mdl-23550644

ABSTRACT

MicroRNAs (miRNAs) are a newly recognized class of regulatory genes which repress the expression of protein-coding genes. Numerous studies have uncovered a complex role for miRNAs regulating many aspects of a variety of cellular processes including cell growth, differentiation, and lineage commitment. In the immune system, miR-155 is unique in its ability to shape the transcriptome of activated myeloid and lymphoid cells controlling diverse biological functions ranging from inflammation to immunological memory. Not surprisingly, a tight control of miR-155 expression is required to avoid malignant transformation, as evidenced by miR-155 overexpression in many cancers of B-cell origin. In this review, we discuss the potential of miR-155 as a molecular target for therapeutic intervention and discuss the function of miR-155 in the context of protective immunity. We first look back into the emergence of miR-155 in evolution, which is coincidental with the emergence of the ancestors of the antigen receptors. We then summarize what we have learned about the role of miR-155 in the regulation of lymphoid subsets at the cellular and molecular level in the context of recent progress in this field.


Subject(s)
B-Lymphocytes/immunology , Immune System/physiology , Lymphoma, B-Cell/immunology , MicroRNAs/immunology , Myeloid Cells/immunology , Animals , Biological Evolution , Cell Differentiation , Cell Lineage , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/immunology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunologic Memory , Lymphoma, B-Cell/genetics , MicroRNAs/genetics , Receptors, Antigen/genetics , Transcriptome
15.
Infect Immun ; 81(3): 723-32, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23264052

ABSTRACT

MicroRNAs (miRNAs) are small noncoding molecules that control gene expression posttranscriptionally, with microRNA-155 (miR-155) one of the first to be implicated in immune regulation. Here, we show that miR-155-deficient mice are less able to eradicate a mucosal Citrobacter rodentium infection than wild-type C57BL/6 mice. miR-155-deficient mice exhibited prolonged colonization associated with a higher C. rodentium burden in gastrointestinal tissue and spread into systemic tissues. Germinal center formation and humoral immune responses against C. rodentium were severely impaired in infected miR-155-deficient mice. A similarly susceptible phenotype was observed in µMT mice reconstituted with miR-155-deficient B cells, indicating that miR-155 is required intrinsically for mediating protection against this predominantly luminal bacterial pathogen.


Subject(s)
Citrobacter rodentium , Enterobacteriaceae Infections/microbiology , Genetic Predisposition to Disease , MicroRNAs/metabolism , Animals , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/immunology , Female , Gene Expression Regulation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Real-Time Polymerase Chain Reaction , Transcriptome
16.
PLoS One ; 7(8): e40938, 2012.
Article in English | MEDLINE | ID: mdl-22912667

ABSTRACT

BACKGROUND: miRNAs are a class of small non-coding RNAs that regulate gene expression and have critical functions in various biological processes. Hundreds of miRNAs have been identified in mammalian genomes but only a small number of them have been functionally characterized. Recent studies also demonstrate that some miRNAs have important roles in reprogramming somatic cells to induced pluripotent stem cells (iPSCs). METHODS: We screened 52 miRNAs cloned in a piggybac (PB) vector for their roles in reprogramming of mouse embryonic fibroblast cells to iPSCs. To identify targets of miRNAs, we made Dgcr8-deficient embryonic stem (ES) cells and introduced miRNA mimics to these cells, which lack miRNA biogenesis. The direct target genes of miRNA were identified through global gene expression analysis and target validation. RESULTS AND CONCLUSION: We found that over-expressing miR-25 or introducing miR-25 mimics enhanced production of iPSCs. We identified a number of miR-25 candidate gene targets. Of particular interest were two ubiquitin ligases, Wwp2 and Fbxw7, which have been proposed to regulate Oct4, c-Myc and Klf5, respectively. Our findings thus highlight the complex interplay between miRNAs and transcription factors involved in reprogramming, stem cell self-renewal and maintenance of pluripotency.


Subject(s)
Cellular Reprogramming/genetics , F-Box Proteins/genetics , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/cytology , MicroRNAs/genetics , MicroRNAs/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Base Sequence , Computational Biology , F-Box-WD Repeat-Containing Protein 7 , Fibroblasts/cytology , Gene Expression Regulation , Mice
17.
Nucleic Acids Res ; 40(5): 2152-67, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22070883

ABSTRACT

Anti-miRs are oligonucleotide inhibitors complementary to miRNAs that have been used extensively as tools to gain understanding of specific miRNA functions and as potential therapeutics. We showed previously that peptide nucleic acid (PNA) anti-miRs containing a few attached Lys residues were potent miRNA inhibitors. Using miR-122 as an example, we report here the PNA sequence and attached amino acid requirements for efficient miRNA targeting and show that anti-miR activity is enhanced substantially by the presence of a terminal-free thiol group, such as a Cys residue, primarily due to better cellular uptake. We show that anti-miR activity of a Cys-containing PNA is achieved by cell uptake through both clathrin-dependent and independent routes. With the aid of two PNA analogues having intrinsic fluorescence, thiazole orange (TO)-PNA and [bis-o-(aminoethoxy)phenyl]pyrrolocytosine (BoPhpC)-PNA, we explored the subcellular localization of PNA anti-miRs and our data suggest that anti-miR targeting of miR-122 may take place in or associated with endosomal compartments. Our findings are valuable for further design of PNAs and other oligonucleotides as potent anti-miR agents.


Subject(s)
MicroRNAs/antagonists & inhibitors , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/metabolism , Amino Acids/chemistry , Cell Line , Endocytosis , Endosomes , Humans , Peptide Nucleic Acids/analysis , Sulfhydryl Compounds/chemistry , Up-Regulation
18.
RNA ; 17(5): 933-43, 2011 May.
Article in English | MEDLINE | ID: mdl-21441346

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs involved in fine-tuning of gene regulation. Antisense oligonucleotides (ONs) are promising tools as anti-miRNA (anti-miR) agents toward therapeutic applications and to uncover miRNA function. Such anti-miR ONs include 2'-O-methyl (OMe), cationic peptide nucleic acids like K-PNA-K3, and locked nucleic acid (LNA)-based anti-miRs such as LNA/DNA or LNA/OMe. Northern blotting is a widely used and robust technique to detect miRNAs. However, miRNA quantification in the presence of anti-miR ONs has proved to be challenging, due to detection artifacts, which has led to poor understanding of miRNA fate upon anti-miR binding. Here we show that anti-miR ON bound to miR-122 can prevent the miRNA from being properly precipitated into the purified RNA fraction using the standard RNA extraction protocol (TRI-Reagent), yielding an RNA extract that does not reflect the real cellular levels of the miRNA. An increase in the numbers of equivalents of isopropanol during the precipitation step leads to full recovery of the targeted miRNA back into the purified RNA extract. Following our improved protocol, we demonstrate by Northern blotting, in conjunction with a PNA decoy strategy and use of high denaturing PAGE, that high-affinity anti-miRs (K-PNA-K3, LNA/DNA, and LNA/OMe) sequester miR-122 without causing miRNA degradation, while miR-122 targeting with a lower-affinity anti-miR (OMe) seems to promote degradation of the miRNA. The technical issues explored in this work will have relevance for other hybridization-based techniques for miRNA quantification in the presence of anti-miR ONs.


Subject(s)
Blotting, Northern/methods , MicroRNAs/analysis , Oligonucleotides/analysis , Cell Line, Tumor , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Oligonucleotides/genetics , RNA Stability
19.
Nucleic Acids Res ; 38(13): 4466-75, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20223773

ABSTRACT

MicroRNAs (miRNAs) play an important role in diverse physiological processes and are potential therapeutic agents. Synthetic oligonucleotides (ONs) of different chemistries have proven successful for blocking miRNA expression. However, their specificity and efficiency have not been fully evaluated. Here, we show that peptide nucleic acids (PNAs) efficiently block a key inducible miRNA expressed in the haematopoietic system, miR-155, in cultured B cells as well as in mice. Remarkably, miR-155 inhibition by PNA in primary B cells was achieved in the absence of any transfection agent. In mice, the high efficiency of the treatment was demonstrated by a strong overlap in global gene expression between B cells isolated from anti-miR-155 PNA-treated and miR-155-deficient mice. Interestingly, PNA also induced additional changes in gene expression. Our analysis provides a useful platform to aid the design of efficient and specific anti-miRNA ONs for in vivo use.


Subject(s)
Gene Expression Regulation , MicroRNAs/antagonists & inhibitors , Peptide Nucleic Acids , Animals , B-Lymphocytes/metabolism , Binding Sites , Cells, Cultured , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Microwaves , Peptide Nucleic Acids/chemical synthesis , RNA, Messenger/chemistry , RNA, Messenger/metabolism
20.
Nat Immunol ; 11(3): 265-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20139987

ABSTRACT

Voltage-gated proton currents regulate generation of reactive oxygen species (ROS) in phagocytic cells. In B cells, stimulation of the B cell antigen receptor (BCR) results in the production of ROS that participate in B cell activation, but the involvement of proton channels is unknown. We report here that the voltage-gated proton channel HVCN1 associated with the BCR complex and was internalized together with the BCR after activation. BCR-induced generation of ROS was lower in HVCN1-deficient B cells, which resulted in attenuated BCR signaling via impaired BCR-dependent oxidation of the tyrosine phosphatase SHP-1. This resulted in less activation of the kinases Syk and Akt, impaired mitochondrial respiration and glycolysis and diminished antibody responses in vivo. Our findings identify unanticipated functions for proton channels in B cells and demonstrate the importance of ROS in BCR signaling and downstream metabolism.


Subject(s)
B-Lymphocytes/immunology , Ion Channels/immunology , Reactive Oxygen Species/immunology , Receptors, Antigen, B-Cell/immunology , Animals , B-Lymphocytes/enzymology , Enzyme Activation/immunology , Immunoblotting , Intracellular Signaling Peptides and Proteins/immunology , Mice , Mice, Knockout , Microscopy, Confocal , Mitochondria/immunology , Oncogene Protein v-akt/immunology , Protein-Tyrosine Kinases/immunology , Signal Transduction , Syk Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...