Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Genome Res ; 23(7): 1130-41, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23570689

ABSTRACT

Nearly three-quarters of the 143 genetic signals associated with platelet and erythrocyte phenotypes identified by meta-analyses of genome-wide association (GWA) studies are located at non-protein-coding regions. Here, we assessed the role of candidate regulatory variants associated with cell type-restricted, closely related hematological quantitative traits in biologically relevant hematopoietic cell types. We used formaldehyde-assisted isolation of regulatory elements followed by next-generation sequencing (FAIRE-seq) to map regions of open chromatin in three primary human blood cells of the myeloid lineage. In the precursors of platelets and erythrocytes, as well as in monocytes, we found that open chromatin signatures reflect the corresponding hematopoietic lineages of the studied cell types and associate with the cell type-specific gene expression patterns. Dependent on their signal strength, open chromatin regions showed correlation with promoter and enhancer histone marks, distance to the transcription start site, and ontology classes of nearby genes. Cell type-restricted regions of open chromatin were enriched in sequence variants associated with hematological indices. The majority (63.6%) of such candidate functional variants at platelet quantitative trait loci (QTLs) coincided with binding sites of five transcription factors key in regulating megakaryopoiesis. We experimentally tested 13 candidate regulatory variants at 10 platelet QTLs and found that 10 (76.9%) affected protein binding, suggesting that this is a frequent mechanism by which regulatory variants influence quantitative trait levels. Our findings demonstrate that combining large-scale GWA data with open chromatin profiles of relevant cell types can be a powerful means of dissecting the genetic architecture of closely related quantitative traits.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Genetic Variation , Quantitative Trait Loci , Quantitative Trait, Heritable , Regulatory Sequences, Nucleic Acid , Blood Platelets/metabolism , Cell Lineage/genetics , Chromosome Mapping , Cluster Analysis , Erythrocytes/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Histones/metabolism , Humans , Myeloid Cells/metabolism , Nucleosomes/metabolism , Organ Specificity/genetics , Phenotype , Polymorphism, Single Nucleotide
2.
Nature ; 492(7429): 369-75, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23222517

ABSTRACT

Anaemia is a chief determinant of global ill health, contributing to cognitive impairment, growth retardation and impaired physical capacity. To understand further the genetic factors influencing red blood cells, we carried out a genome-wide association study of haemoglobin concentration and related parameters in up to 135,367 individuals. Here we identify 75 independent genetic loci associated with one or more red blood cell phenotypes at P < 10(-8), which together explain 4-9% of the phenotypic variance per trait. Using expression quantitative trait loci and bioinformatic strategies, we identify 121 candidate genes enriched in functions relevant to red blood cell biology. The candidate genes are expressed preferentially in red blood cell precursors, and 43 have haematopoietic phenotypes in Mus musculus or Drosophila melanogaster. Through open-chromatin and coding-variant analyses we identify potential causal genetic variants at 41 loci. Our findings provide extensive new insights into genetic mechanisms and biological pathways controlling red blood cell formation and function.


Subject(s)
Erythrocytes/metabolism , Genetic Loci , Genome-Wide Association Study , Phenotype , Animals , Cell Cycle/genetics , Cytokines/metabolism , Drosophila melanogaster/genetics , Erythrocytes/cytology , Female , Gene Expression Regulation/genetics , Hematopoiesis/genetics , Hemoglobins/genetics , Humans , Male , Mice , Organ Specificity , Polymorphism, Single Nucleotide/genetics , RNA Interference , Signal Transduction/genetics
3.
Blood ; 120(24): 4859-68, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22972982

ABSTRACT

We recently identified 68 genomic loci where common sequence variants are associated with platelet count and volume. Platelets are formed in the bone marrow by megakaryocytes, which are derived from hematopoietic stem cells by a process mainly controlled by transcription factors. The homeobox transcription factor MEIS1 is uniquely transcribed in megakaryocytes and not in the other lineage-committed blood cells. By ChIP-seq, we show that 5 of the 68 loci pinpoint a MEIS1 binding event within a group of 252 MK-overexpressed genes. In one such locus in DNM3, regulating platelet volume, the MEIS1 binding site falls within a region acting as an alternative promoter that is solely used in megakaryocytes, where allelic variation dictates different levels of a shorter transcript. The importance of dynamin activity to the latter stages of thrombopoiesis was confirmed by the observation that the inhibitor Dynasore reduced murine proplatelet for-mation in vitro.


Subject(s)
Blood Platelets/metabolism , Dynamin III/genetics , Genome, Human/genetics , Homeodomain Proteins/genetics , Megakaryocytes/metabolism , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Animals , Binding Sites/genetics , Blood Platelets/drug effects , Cell Line, Tumor , Cell Lineage/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Gene Expression , Genetic Variation , Homeodomain Proteins/metabolism , Humans , Hydrazones/pharmacology , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/metabolism , Platelet Count , Polymorphism, Single Nucleotide , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription Initiation Site , Transcription, Genetic
4.
Nature ; 480(7376): 201-8, 2011 Nov 30.
Article in English | MEDLINE | ID: mdl-22139419

ABSTRACT

Platelets are the second most abundant cell type in blood and are essential for maintaining haemostasis. Their count and volume are tightly controlled within narrow physiological ranges, but there is only limited understanding of the molecular processes controlling both traits. Here we carried out a high-powered meta-analysis of genome-wide association studies (GWAS) in up to 66,867 individuals of European ancestry, followed by extensive biological and functional assessment. We identified 68 genomic loci reliably associated with platelet count and volume mapping to established and putative novel regulators of megakaryopoiesis and platelet formation. These genes show megakaryocyte-specific gene expression patterns and extensive network connectivity. Using gene silencing in Danio rerio and Drosophila melanogaster, we identified 11 of the genes as novel regulators of blood cell formation. Taken together, our findings advance understanding of novel gene functions controlling fate-determining events during megakaryopoiesis and platelet formation, providing a new example of successful translation of GWAS to function.


Subject(s)
Blood Platelets/cytology , Hematopoiesis/genetics , Megakaryocytes/cytology , Animals , Blood Platelets/metabolism , Cell Size , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Europe , Gene Expression Profiling , Gene Silencing , Genome, Human/genetics , Genome-Wide Association Study , Humans , Megakaryocytes/metabolism , Platelet Count , Protein Interaction Maps , Transcription, Genetic/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
5.
Nat Genet ; 43(8): 735-7, 2011 Jul 17.
Article in English | MEDLINE | ID: mdl-21765411

ABSTRACT

Gray platelet syndrome (GPS) is a predominantly recessive platelet disorder that is characterized by mild thrombocytopenia with large platelets and a paucity of α-granules; these abnormalities cause mostly moderate but in rare cases severe bleeding. We sequenced the exomes of four unrelated individuals and identified NBEAL2 as the causative gene; it has no previously known function but is a member of a gene family that is involved in granule development. Silencing of nbeal2 in zebrafish abrogated thrombocyte formation.


Subject(s)
Blood Platelets/metabolism , Cytoplasmic Granules/metabolism , Gray Platelet Syndrome/genetics , Nerve Tissue Proteins/genetics , Secretory Vesicles/metabolism , Adult , Aged , Animals , Animals, Genetically Modified , Base Sequence , Blood Platelets/pathology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Female , Gene Expression Regulation, Developmental , Humans , Male , Middle Aged , Molecular Sequence Data , Nerve Tissue Proteins/antagonists & inhibitors , Pedigree , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Young Adult , Zebrafish/growth & development , Zebrafish/metabolism
6.
Hum Mutat ; 30(6): 1003-11, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19370760

ABSTRACT

Cerebral cavernous malformations (CCMs) may cause recurrent headaches, seizures, and hemorrhagic stroke and have been associated with loss-of-function mutations in CCM1/KRIT1, CCM2, and CCM3/programmed cell death 10 (PDCD10). The CCM3/PDCD10 amino acid sequence does not reveal significant homologies to protein domains with known structure. With the help of the only published human in-frame deletion of the CCM3 gene (c.97-?_150+?del), CCM3:p.L33_K50del, we have identified the interaction domain of CCM3 with the oxidant stress response serine/threonine kinase 25 (STK25, YSK1, SOK1) and with the mammalian Ste20-like kinase 4 (MST4, MASK). Consistently, nano-liquid chromatography-tandem mass spectrometry (nano-LC-MS/MS) analyses revealed two STK25 phosphorylation sites at serine 39 and threonine 43. The corresponding in-frame deletion of zebrafish ccm3a, dccm3:p.L31_K48del, also resulted in impaired interaction with STK25 and MST4. In agreement with the observed redundant biochemical functionality of zebrafish ccm3a and its duplicate ccm3b, simultaneous inactivation of both genes resulted in a progressive cardiovascular phenotype in zebrafish indistinguishable from ccm1 and ccm2 mutants. The pronounced cardiovascular dilatations could be recapitulated by morpholino-induced in-frame skipping of the exon encoding the STK25 and MST4 binding site of zebrafish Ccm3a if Ccm3b was repressed in parallel. Using a novel zebrafish model of CCM, we could thus demonstrate that the newly mapped STK25 and MST4 interaction domain within the CCM3 protein plays a crucial role for vascular development in zebrafish.


Subject(s)
Amino Acids/genetics , Apoptosis Regulatory Proteins/genetics , Frameshift Mutation/genetics , Membrane Proteins/genetics , Protein Interaction Mapping , Proto-Oncogene Proteins/genetics , Sequence Deletion , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Cardiovascular Abnormalities/embryology , Cardiovascular Abnormalities/metabolism , Carrier Proteins/metabolism , Cell Line , Exons/genetics , Gene Knockdown Techniques , Heart/embryology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , KRIT1 Protein , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Phenotype , Phosphorylation , Phosphoserine/metabolism , Phosphothreonine/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Zebrafish/embryology , Zebrafish Proteins/chemistry
7.
Nat Genet ; 40(3): 329-34, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18297070

ABSTRACT

Hypotrichosis simplex is a group of nonsyndromic human alopecias. We mapped an autosomal recessive form of this disorder to chromosome 13q14.11-13q21.33, and identified homozygous truncating mutations in P2RY5, which encodes an orphan G protein-coupled receptor. Furthermore, we identified oleoyl-L-alpha-lysophosphatidic acid (LPA), a bioactive lipid, as a ligand for P2Y5 in reporter gene and radioligand binding experiments. Homology and studies of signaling transduction pathways suggest that P2Y5 is a member of a subgroup of LPA receptors, which also includes LPA4 and LPA5. Our study is the first to implicate a G protein-coupled receptor as essential for and specific to the maintenance of human hair growth. This finding may provide opportunities for new therapeutic approaches to the treatment of hair loss in humans.


Subject(s)
Hair/growth & development , Hypotrichosis/genetics , Lysophospholipids/physiology , Receptors, Purinergic P2/physiology , Adult , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Chromosome Mapping , Chromosomes, Human, Pair 13 , Consanguinity , DNA Mutational Analysis , Family , Humans , Lysophospholipids/metabolism , Models, Biological , Pedigree , Phylogeny , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Transfection
8.
Hum Mutat ; 29(5): 709-17, 2008 May.
Article in English | MEDLINE | ID: mdl-18300272

ABSTRACT

Cerebral cavernous malformations (CCM) are prevalent cerebrovascular lesions predisposing to chronic headaches, epilepsy, and hemorrhagic stroke. Using a combination of direct sequencing and MLPA analyses, we identified 15 novel and eight previously published CCM1 (KRIT1), CCM2, and CCM3 (PDCD10) mutations. The mutation detection rate was >90% for familial cases and >60% for isolated cases with multiple malformations. Splice site mutations constituted almost 20% of all CCM mutations identified. One of these proved to be a de novo mutation of the most 3' acceptor splice site of the CCM1 gene resulting in retention of intron 19. A further mutation affected the 3' splice site of CCM2 intron 2 leading to cryptic splice site utilization in both CCM2 and its transcript variant lacking exon 2. With the exception of one in-frame deletion of CCM2 exon 2, which corresponds to the naturally occurring splice variant of CCM2 on the RNA level and is predicted to result in the omission of 58 amino acids (CCM2:p.P11_K68del), all mutations lead to the introduction of premature stop codons. To gain insight into the likely mechanisms underlying the only known CCM2 in-frame deletion, we analyzed the functional consequences of loss of CCM2 exon 2. The CCM2:p.P11_K68del protein could be expressed in cell culture and complexed with CCM3. However, its ability to interact with CCM1 and to form a CCM1/CCM2/CCM3 complex was lost. These data are in agreement with a loss-of-function mechanism for CCM mutations, uncover an N-terminal CCM2 domain required for CCM1 binding, and demonstrate full-length CCM2 as the essential core protein in the CCM1/CCM2/CCM3 complex.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carrier Proteins/genetics , Hemangioma, Cavernous, Central Nervous System/genetics , Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Mutation , Proto-Oncogene Proteins/genetics , Sequence Deletion , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Carrier Proteins/metabolism , Cell Line , DNA Primers , Female , Humans , KRIT1 Protein , Male , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Pedigree , Protein Binding , Proto-Oncogene Proteins/metabolism
9.
Neurogenetics ; 8(4): 249-56, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17657516

ABSTRACT

Individuals carrying a mutation in one of the three cerebral cavernous malformation genes (CCM1/KRIT1, CCM2, CCM3) cannot be clinically distinguished, raising the possibility that they act within common molecular pathways. In this study, we demonstrate that CCM3 (PDCD10) coprecipitates and colocalizes with CCM2. We also show that CCM3 directly binds to serine/threonine kinase 25 (STK25, YSK1, SOK1) and the phosphatase domain of Fas-associated phosphatase-1 (FAP-1, PTPN13, PTP-Bas, PTP-BL). CCM3 is phosphorylated by STK25 but not by its other Yeast-Two hybrid interactor STK24, whereas the C-terminal catalytic domain of FAP-1 dephosphorylates CCM3. Finally, our experiments reveal that STK25 forms a protein complex with CCM2. Thus, our data link two proteins of unknown function, CCM3 and STK25, with CCM2, which is part of signaling pathways essential for vascular development and CCM pathogenesis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carrier Proteins/genetics , Hemangioma, Cavernous, Central Nervous System/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Amino Acid Sequence , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Line , Germ-Line Mutation , Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/metabolism , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Protein Tyrosine Phosphatase, Non-Receptor Type 13/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Two-Hybrid System Techniques
10.
Eur J Immunol ; 34(5): 1405-13, 2004 May.
Article in English | MEDLINE | ID: mdl-15114674

ABSTRACT

Notch receptors are involved in directing the choice between alternative cell fates in developmental scenarios such as thymopoiesis. By pharmacological interference in rat fetal thymus organ culture we show that inhibition of Notch signaling arrests T cell development at an early double-negative stage and is accompanied by a dramatic increase in the number of NK cells. These cells show an activated phenotype, lack recombination of the TCR beta gene locus and express perforin. Similarly, in thymic lobes reconstituted with fetal liver cells, progenitors predominantly develop into NK cells both after pharmacological interference of Notch and after treatment with a recombinant rat Notch1/Fc chimera. Collectively, this identifies the lineage decision of NK/T precursor cells as an important site of Notch action in rat thymocytes.


Subject(s)
Cell Differentiation/physiology , Killer Cells, Natural/metabolism , Membrane Proteins/metabolism , Thymus Gland/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , Amyloid Precursor Protein Secretases , Animals , Antigens, Surface/metabolism , Endopeptidases/drug effects , Lectins, C-Type/metabolism , Liver/embryology , Liver/physiology , Membrane Proteins/drug effects , NK Cell Lectin-Like Receptor Subfamily B , Rats , Receptors, Notch , Signal Transduction/drug effects , Signal Transduction/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Triglycerides/pharmacology , gamma-Aminobutyric Acid/pharmacology
11.
J Exp Biol ; 207(Pt 3): 443-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14691092

ABSTRACT

This study on common marmosets Callithrix jacchus is the first to examine noise-dependent mechanisms of vocal plasticity in a New World monkey. Since acoustic communication can be considerably impaired by environmental noise, some animals have evolved adaptations to counteract its masking effects. The studied marmosets increased the sound level of their spontaneous calls in response to increased levels of white noise broadcast to them. Possibly, such noise-dependent adjustment of vocal amplitude serves to maintain a specific signal-to-noise ratio that is favourable for signal production. Concurrently, the adjustment of vocal amplitude can maintain a given active space for communication. In contrast to some bird species, no noise-induced increase in the number of syllables per call series could be found, showing that an increased serial redundancy of vocal signals was not used to communicate under noisy conditions. Finally, we examined a possible noise-dependent prolongation of vocal signals. This approach was guided by the findings of perceptional studies, which suggest an increased detection probability of prolonged signals in noise by temporal summation. Marmosets indeed increased the duration of their call syllables along with increasing background noise levels. This is the first evidence of such mechanism of vocal plasticity in an animal communication system.


Subject(s)
Adaptation, Physiological , Callithrix/physiology , Noise , Vocalization, Animal/physiology , Animals , Sound Spectrography , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...