Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Euro Surveill ; 29(23)2024 Jun.
Article in English | MEDLINE | ID: mdl-38847119

ABSTRACT

BackgroundThe COVID-19 pandemic was largely driven by genetic mutations of SARS-CoV-2, leading in some instances to enhanced infectiousness of the virus or its capacity to evade the host immune system. To closely monitor SARS-CoV-2 evolution and resulting variants at genomic-level, an innovative pipeline termed SARSeq was developed in Austria.AimWe discuss technical aspects of the SARSeq pipeline, describe its performance and present noteworthy results it enabled during the pandemic in Austria.MethodsThe SARSeq pipeline was set up as a collaboration between private and public clinical diagnostic laboratories, a public health agency, and an academic institution. Representative SARS-CoV-2 positive specimens from each of the nine Austrian provinces were obtained from SARS-CoV-2 testing laboratories and processed centrally in an academic setting for S-gene sequencing and analysis.ResultsSARS-CoV-2 sequences from up to 2,880 cases weekly resulted in 222,784 characterised case samples in January 2021-March 2023. Consequently, Austria delivered the fourth densest genomic surveillance worldwide in a very resource-efficient manner. While most SARS-CoV-2 variants during the study showed comparable kinetic behaviour in all of Austria, some, like Beta, had a more focused spread. This highlighted multifaceted aspects of local population-level acquired immunity. The nationwide surveillance system enabled reliable nowcasting. Measured early growth kinetics of variants were predictive of later incidence peaks.ConclusionWith low automation, labour, and cost requirements, SARSeq is adaptable to monitor other pathogens and advantageous even for resource-limited countries. This multiplexed genomic surveillance system has potential as a rapid response tool for future emerging threats.


Subject(s)
COVID-19 , Genome, Viral , SARS-CoV-2 , Humans , Austria/epidemiology , SARS-CoV-2/genetics , COVID-19/epidemiology , COVID-19/virology , COVID-19/diagnosis , Mutation , Genomics/methods , Pandemics , Evolution, Molecular , Whole Genome Sequencing/methods
2.
EMBO J ; 40(22): e108234, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34586646

ABSTRACT

DNA methylation is a fundamental epigenetic modification, important across biological processes. The maintenance methyltransferase DNMT1 is essential for lineage differentiation during development, but its functions in tissue homeostasis are incompletely understood. We show that epidermis-specific DNMT1 deletion severely disrupts epidermal structure and homeostasis, initiating a massive innate immune response and infiltration of immune cells. Mechanistically, DNA hypomethylation in keratinocytes triggered transposon derepression, mitotic defects, and formation of micronuclei. DNA release into the cytosol of DNMT1-deficient keratinocytes activated signaling through cGAS and STING, thus triggering inflammation. Our findings show that disruption of a key epigenetic mark directly impacts immune and tissue homeostasis, and potentially impacts our understanding of autoinflammatory diseases and cancer immunotherapy.


Subject(s)
DNA Methylation , Dermatitis/genetics , Epidermis/physiopathology , Nucleotidyltransferases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chromosome Aberrations , Cytosol/physiology , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Dermatitis/immunology , Dermatitis/pathology , Humans , Immunity, Innate/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Keratinocytes/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Transgenic , Nucleotidyltransferases/genetics
3.
Genome Res ; 24(11): 1808-20, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25135956

ABSTRACT

The selectivity of transcriptional responses to extracellular cues is reflected by the deposition of stimulus-specific chromatin marks. Although histone H3 phosphorylation is a target of numerous signaling pathways, its role in transcriptional regulation remains poorly understood. Here, for the first time, we report a genome-wide analysis of H3S28 phosphorylation in a mammalian system in the context of stress signaling. We found that this mark targets as many as 50% of all stress-induced genes, underlining its importance in signal-induced transcription. By combining ChIP-seq, RNA-seq, and mass spectrometry we identified the factors involved in the biological interpretation of this histone modification. We found that MSK1/2-mediated phosphorylation of H3S28 at stress-responsive promoters contributes to the dissociation of HDAC corepressor complexes and thereby to enhanced local histone acetylation and subsequent transcriptional activation of stress-induced genes. Our data reveal a novel function of the H3S28ph mark in the activation of mammalian genes in response to MAP kinase pathway activation.


Subject(s)
Histones/metabolism , Serine/metabolism , Stress, Physiological/genetics , Transcriptional Activation , 3T3 Cells , Acetylation , Animals , Chromatin Immunoprecipitation , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Ontology , Genome-Wide Association Study , HeLa Cells , High-Throughput Nucleotide Sequencing , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , MAP Kinase Signaling System/genetics , Mice , Oligonucleotide Array Sequence Analysis , Phosphorylation , Promoter Regions, Genetic/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
4.
Immunity ; 39(2): 229-44, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973221

ABSTRACT

The immunoglobulin heavy-chain (Igh) locus undergoes large-scale contraction in pro-B cells, which facilitates VH-DJH recombination by juxtaposing distal VH genes next to the DJH-rearranged gene segment in the 3' proximal Igh domain. By using high-resolution mapping of long-range interactions, we demonstrate that local interaction domains established the three-dimensional structure of the extended Igh locus in lymphoid progenitors. In pro-B cells, these local domains engaged in long-range interactions across the Igh locus, which depend on the regulators Pax5, YY1, and CTCF. The large VH gene cluster underwent flexible long-range interactions with the more rigidly structured proximal domain, which probably ensures similar participation of all VH genes in VH-DJH recombination to generate a diverse antibody repertoire. These long-range interactions appear to be an intrinsic feature of the VH gene cluster, because they are still generated upon mutation of the Eµ enhancer, IGCR1 insulator, or 3' regulatory region in the proximal Igh domain.


Subject(s)
Antibody Diversity/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain/genetics , Genes, Immunoglobulin Heavy Chain , Immunoglobulin Variable Region/genetics , Precursor Cells, B-Lymphoid/immunology , Animals , Base Sequence , Binding Sites , CCCTC-Binding Factor , Chromosome Mapping , Gene Rearrangement , Mice , Mice, Inbred C57BL , PAX5 Transcription Factor/metabolism , Protein Binding , Repressor Proteins/metabolism , Sequence Analysis, DNA , YY1 Transcription Factor/metabolism
5.
Nature ; 495(7442): 474-80, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23474986

ABSTRACT

CLP1 was the first mammalian RNA kinase to be identified. However, determining its in vivo function has been elusive. Here we generated kinase-dead Clp1 (Clp1(K/K)) mice that show a progressive loss of spinal motor neurons associated with axonal degeneration in the peripheral nerves and denervation of neuromuscular junctions, resulting in impaired motor function, muscle weakness, paralysis and fatal respiratory failure. Transgenic rescue experiments show that CLP1 functions in motor neurons. Mechanistically, loss of CLP1 activity results in accumulation of a novel set of small RNA fragments, derived from aberrant processing of tyrosine pre-transfer RNA. These tRNA fragments sensitize cells to oxidative-stress-induced p53 (also known as TRP53) activation and p53-dependent cell death. Genetic inactivation of p53 rescues Clp1(K/K) mice from the motor neuron loss, muscle denervation and respiratory failure. Our experiments uncover a mechanistic link between tRNA processing, formation of a new RNA species and progressive loss of lower motor neurons regulated by p53.


Subject(s)
Motor Neurons/metabolism , Motor Neurons/pathology , RNA, Transfer, Tyr/metabolism , Transcription Factors/metabolism , Amyotrophic Lateral Sclerosis , Animals , Animals, Newborn , Axons/metabolism , Axons/pathology , Cell Death , Diaphragm/innervation , Embryo Loss , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Exons/genetics , Female , Fibroblasts , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscular Atrophy, Spinal , Neuromuscular Diseases/metabolism , Neuromuscular Diseases/pathology , Oxidative Stress , RNA Processing, Post-Transcriptional , RNA, Transfer, Tyr/genetics , RNA-Binding Proteins , Respiration , Spinal Nerves/cytology , Transcription Factors/deficiency , Tumor Suppressor Protein p53/metabolism , Tyrosine/genetics , Tyrosine/metabolism
6.
EMBO J ; 31(14): 3130-46, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22669466

ABSTRACT

Pax5 controls the identity and development of B cells by repressing lineage-inappropriate genes and activating B-cell-specific genes. Here, we used genome-wide approaches to identify Pax5 target genes in pro-B and mature B cells. In these cell types, Pax5 bound to 40% of the cis-regulatory elements defined by mapping DNase I hypersensitive (DHS) sites, transcription start sites and histone modifications. Although Pax5 bound to 8000 target genes, it regulated only 4% of them in pro-B and mature B cells by inducing enhancers at activated genes and eliminating DHS sites at repressed genes. Pax5-regulated genes in pro-B cells account for 23% of all expression changes occurring between common lymphoid progenitors and committed pro-B cells, which identifies Pax5 as an important regulator of this developmental transition. Regulated Pax5 target genes minimally overlap in pro-B and mature B cells, which reflects massive expression changes between these cell types. Hence, Pax5 controls B-cell identity and function by regulating distinct target genes in early and late B lymphopoiesis.


Subject(s)
Gene Expression Regulation/physiology , Lymphopoiesis/physiology , PAX5 Transcription Factor/metabolism , Precursor Cells, B-Lymphoid/metabolism , Response Elements/physiology , Transcription, Genetic/physiology , Animals , Mice , PAX5 Transcription Factor/genetics , Precursor Cells, B-Lymphoid/cytology
7.
PLoS One ; 6(11): e27288, 2011.
Article in English | MEDLINE | ID: mdl-22102886

ABSTRACT

Imprinted macro non-protein-coding (nc) RNAs are cis-repressor transcripts that silence multiple genes in at least three imprinted gene clusters in the mouse genome. Similar macro or long ncRNAs are abundant in the mammalian genome. Here we present the full coding and non-coding transcriptome of two mouse tissues: differentiated ES cells and fetal head using an optimized RNA-Seq strategy. The data produced is highly reproducible in different sequencing locations and is able to detect the full length of imprinted macro ncRNAs such as Airn and Kcnq1ot1, whose length ranges between 80-118 kb. Transcripts show a more uniform read coverage when RNA is fragmented with RNA hydrolysis compared with cDNA fragmentation by shearing. Irrespective of the fragmentation method, all coding and non-coding transcripts longer than 8 kb show a gradual loss of sequencing tags towards the 3' end. Comparisons to published RNA-Seq datasets show that the strategy presented here is more efficient in detecting known functional imprinted macro ncRNAs and also indicate that standardization of RNA preparation protocols would increase the comparability of the transcriptome between different RNA-Seq datasets.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genomic Imprinting , Head/physiology , RNA, Untranslated/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Fetus , Gene Expression Profiling , Genome , Mice , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
8.
EMBO J ; 30(12): 2388-404, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21552207

ABSTRACT

Pax5 is a critical regulator of B-cell commitment. Here, we identified direct Pax5 target genes by streptavidin-mediated ChIP-chip analysis of pro-B cells expressing in vivo biotinylated Pax5. By binding to promoters and enhancers, Pax5 directly regulates the expression of multiple transcription factor, cell surface receptor and signal transducer genes. One of the newly identified enhancers was shown by transgenic analysis to confer Pax5-dependent B-cell-specific activity to the Nedd9 gene controlling B-cell trafficking. Profiling of histone modifications in Pax5-deficient and wild-type pro-B cells demonstrated that Pax5 induces active chromatin at activated target genes, while eliminating active chromatin at repressed genes in committed pro-B cells. Pax5 rapidly induces these chromatin and transcription changes by recruiting chromatin-remodelling, histone-modifying and basal transcription factor complexes to its target genes. These data provide novel insight into the regulatory network and epigenetic regulation, by which Pax5 controls B-cell commitment.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Cell Differentiation , Chromatin/metabolism , Gene Targeting , PAX5 Transcription Factor/physiology , Animals , Cell Differentiation/genetics , Cell Line , Gene Knock-In Techniques , Gene Targeting/methods , Mice , Mice, Knockout , Mice, Transgenic , PAX5 Transcription Factor/genetics , Protein Binding/genetics , Protein Transport/genetics , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism
9.
Immunity ; 34(2): 175-87, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21349430

ABSTRACT

V(H)-DJ(H) recombination of the immunoglobulin heavy chain (Igh) locus is temporally and spatially controlled during early B cell development, and yet no regulatory elements other than the V(H) gene promoters have been identified throughout the entire V(H) gene cluster. Here, we discovered regulatory sequences that are interspersed in the distal V(H) gene region. These conserved repeat elements were characterized by the presence of Pax5 transcription factor-dependent active chromatin by binding of the regulators Pax5, E2A, CTCF, and Rad21, as well as by Pax5-dependent antisense transcription in pro-B cells. The Pax5-activated intergenic repeat (PAIR) elements were no longer bound by Pax5 in pre-B and B cells consistent with the loss of antisense transcription, whereas E2A and CTCF interacted with PAIR elements throughout early B cell development. The pro-B cell-specific and Pax5-dependent activity of the PAIR elements suggests that they are involved in the regulation of distal V(H)-DJ(H) recombination at the Igh locus.


Subject(s)
Chromatin/genetics , DNA, Intergenic/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin/genetics , Immunoglobulin Heavy Chains/genetics , PAX5 Transcription Factor/physiology , Regulatory Sequences, Nucleic Acid/genetics , Animals , B-Lymphocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Binding Sites , CCCTC-Binding Factor , Chromatin Immunoprecipitation , Conserved Sequence , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Genetic , Oligonucleotide Array Sequence Analysis , PAX5 Transcription Factor/deficiency , PAX5 Transcription Factor/genetics , Precursor Cells, B-Lymphoid/metabolism , RNA, Antisense/biosynthesis , RNA, Antisense/genetics , Repressor Proteins/physiology , Transcription, Genetic
10.
Genome Res ; 19(2): 221-33, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19047520

ABSTRACT

In mammals, genome-wide chromatin maps and immunofluorescence studies show that broad domains of repressive histone modifications are present on pericentromeric and telomeric repeats and on the inactive X chromosome. However, only a few autosomal loci such as silent Hox gene clusters have been shown to lie in broad domains of repressive histone modifications. Here we present a ChIP-chip analysis of the repressive H3K27me3 histone modification along chr 17 in mouse embryonic fibroblast cells using an algorithm named broad local enrichments (BLOCs), which allows the identification of broad regions of histone modifications. Our results, confirmed by BLOC analysis of a whole genome ChIP-seq data set, show that the majority of H3K27me3 modifications form BLOCs rather than focal peaks. H3K27me3 BLOCs modify silent genes of all types, plus flanking intergenic regions and their distribution indicates a negative correlation between H3K27me3 and transcription. However, we also found that some nontranscribed gene-poor regions lack H3K27me3. We therefore performed a low-resolution analysis of whole mouse chr 17, which revealed that H3K27me3 is enriched in mega-base-pair-sized domains that are also enriched for genes, short interspersed elements (SINEs) and active histone modifications. These genic H3K27me3 domains alternate with similar-sized gene-poor domains. These are deficient in active histone modifications, as well as H3K27me3, but are enriched for long interspersed elements (LINEs) and long-terminal repeat (LTR) transposons and H3K9me3 and H4K20me3. Thus, an autosome can be seen to contain alternating chromatin bands that predominantly separate genes from one retrotransposon class, which could offer unique domains for the specific regulation of genes or the silencing of autonomous retrotransposons.


Subject(s)
Chromosomes, Mammalian/metabolism , DNA, Intergenic/metabolism , Gene Silencing/physiology , Histones/metabolism , Protein Multimerization/physiology , Algorithms , Animals , Chromatin Immunoprecipitation/methods , Chromosome Banding/methods , Chromosomes, Mammalian/chemistry , Histone Methyltransferases , Histone-Lysine N-Methyltransferase , Lysine/metabolism , Methylation , Mice , Models, Biological , Protein Methyltransferases/metabolism , Protein Processing, Post-Translational/physiology , Substrate Specificity
11.
Cancer Cell ; 10(3): 227-39, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16959614

ABSTRACT

Erk/MAPK and TGFbeta signaling cause epithelial to mesenchymal transition (EMT) and metastasis in mouse mammary epithelial cells (EpH4) transformed with oncogenic Ras (EpRas). In trials to unravel underlying mechanisms, expression profiling for EMT-specific genes identified a secreted interleukin-related protein (ILEI), upregulated exclusively at the translational level. Stable overexpression of ILEI in EpH4 and EpRas cells caused EMT, tumor growth, and metastasis, independent of TGFbeta-R signaling and enhanced by Bcl2. RNAi-mediated knockdown of ILEI in EpRas cells before and after EMT (EpRasXT) prevented and reverted TGFbeta-dependent EMT, also abrogating metastasis formation. ILEI is overexpressed and/or altered in intracellular localization in multiple human tumors, an event strongly correlated to invasion/EMT, metastasis formation, and survival in human colon and breast cancer.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Mesenchymal Stem Cells/cytology , Neoplasm Proteins/metabolism , Animals , Cell Differentiation , Cell Line , Cytokines/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/pathology , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , Protein Biosynthesis/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Survival Rate , Time Factors , Transforming Growth Factor beta/metabolism
12.
Oncogene ; 22(46): 7155-69, 2003 Oct 16.
Article in English | MEDLINE | ID: mdl-14562044

ABSTRACT

Epithelial-to-mesenchymal transition (EMT), a switch of polarized epithelial cells to a migratory, fibroblastoid phenotype, is increasingly considered as an important event during malignant tumor progression and metastasis. To identify molecular players involved in EMT and metastasis, we performed expression profiling of a set of combined in vitro/in vivo cellular models, based on clonal, fully polarized mammary epithelial cells. Seven closely related cell pairs were used, which were modified by defined oncogenes and/or external factors and showed specific aspects of epithelial plasticity relevant to cell migration, local invasion and metastasis. Since mRNA levels do not necessarily reflect protein levels in cells, we used an improved expression profiling method based on polysome-bound RNA, suitable to analyse global gene expression on Affymetrix chips. A substantial fraction of all regulated genes was found to be exclusively controlled at the translational level. Furthermore, profiling of the above multiple cell pairs allowed one to identify small numbers of genes by cluster analysis, specifically correlating gene expression with EMT, metastasis, scattering and/or oncogene function. A small set of genes specifically regulated during EMT was identified, including key regulators and signaling pathways involved in cell proliferation, epithelial polarity, survival and trans-differentiation to mesenchymal-like cells with invasive behavior.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial Cells/physiology , Gene Expression Regulation, Neoplastic/genetics , Mesoderm/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Differentiation , Cell Line , Cluster Analysis , Epithelial Cells/pathology , Female , Gene Expression Profiling , Genes, ras , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/pathology , Mesoderm/pathology , Mice , Neoplasm Invasiveness , Polyribosomes/genetics , Protein Biosynthesis , RNA, Messenger/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL