101
|
High-throughput transcription profiling identifies putative epigenetic regulators of hematopoiesis. Blood 2014; 123:e46-57. [PMID: 24671951 DOI: 10.1182/blood-2013-02-483537] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Hematopoietic differentiation is governed by a complex regulatory program controlling the generation of different lineages of blood cells from multipotent hematopoietic stem cells. The transcriptional program that dictates hematopoietic cell fate and differentiation requires an epigenetic memory function provided by a network of epigenetic factors regulating DNA methylation, posttranslational histone modifications, and chromatin structure. Aberrant interactions between epigenetic factors and transcription factors cause perturbations in the blood cell differentiation program that result in various types of hematopoietic disorders. To elucidate the contributions of different epigenetic factors in human hematopoiesis, high-throughput cap analysis of gene expression was used to build transcription profiles of 199 epigenetic factors in a wide range of blood cells. Our epigenetic transcriptome analysis revealed cell type- (eg, HELLS and ACTL6A), lineage- (eg, MLL), and/or leukemia- (eg, CHD2, CBX8, and EPC1) specific expression of several epigenetic factors. In addition, we show that several epigenetic factors use alternative transcription start sites in different cell types. This analysis could serve as a resource for the scientific community for further characterization of the role of these epigenetic factors in blood development.
Collapse
|
102
|
Abstract
Bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) is a transmembrane protein related to the transforming growth factor-β superfamily, and is highly expressed in platelets and endothelial cells. We previously demonstrated its positive role in thrombus formation using a zebrafish thrombosis model. In the present study, we used Bambi-deficient mice and radiation chimeras to evaluate the function of this receptor in the regulation of both hemostasis and thrombosis. We show that Bambi(-/-) and Bambi(+/-) mice exhibit mildly prolonged bleeding times compared with Bambi(+/+) littermates. In addition, using 2 in vivo thrombosis models in mesenterium or cremaster muscle arterioles, we demonstrate that Bambi-deficient mice form unstable thrombi compared with Bambi(+/+) mice. No defects in thrombin generation in Bambi(-/-) mouse plasma could be detected ex vivo. Moreover, the absence of BAMBI had no effect on platelet counts, platelet activation, aggregation, or platelet procoagulant function. Similar to Bambi(-/-) mice, Bambi(-/-) transplanted with Bambi(+/+) bone marrow formed unstable thrombi in the laser-induced thrombosis model that receded more rapidly than thrombi that formed in Bambi(+/+) mice receiving Bambi(-/-) bone marrow transplants. Taken together, these results provide strong evidence for an important role of endothelium rather than platelet BAMBI as a positive regulator of both thrombus formation and stability.
Collapse
|
103
|
Shameer K, Denny JC, Ding K, Jouni H, Crosslin DR, de Andrade M, Chute CG, Peissig P, Pacheco JA, Li R, Bastarache L, Kho AN, Ritchie MD, Masys DR, Chisholm RL, Larson EB, McCarty CA, Roden DM, Jarvik GP, Kullo IJ. A genome- and phenome-wide association study to identify genetic variants influencing platelet count and volume and their pleiotropic effects. Hum Genet 2014; 133:95-109. [PMID: 24026423 PMCID: PMC3880605 DOI: 10.1007/s00439-013-1355-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 08/22/2013] [Indexed: 12/21/2022]
Abstract
Platelets are enucleated cell fragments derived from megakaryocytes that play key roles in hemostasis and in the pathogenesis of atherothrombosis and cancer. Platelet traits are highly heritable and identification of genetic variants associated with platelet traits and assessing their pleiotropic effects may help to understand the role of underlying biological pathways. We conducted an electronic medical record (EMR)-based study to identify common variants that influence inter-individual variation in the number of circulating platelets (PLT) and mean platelet volume (MPV), by performing a genome-wide association study (GWAS). We characterized genetic variants associated with MPV and PLT using functional, pathway and disease enrichment analyses; we assessed pleiotropic effects of such variants by performing a phenome-wide association study (PheWAS) with a wide range of EMR-derived phenotypes. A total of 13,582 participants in the electronic MEdical Records and GEnomic network had data for PLT and 6,291 participants had data for MPV. We identified five chromosomal regions associated with PLT and eight associated with MPV at genome-wide significance (P < 5E-8). In addition, we replicated 20 SNPs [out of 56 SNPs (α: 0.05/56 = 9E-4)] influencing PLT and 22 SNPs [out of 29 SNPs (α: 0.05/29 = 2E-3)] influencing MPV in a published meta-analysis of GWAS of PLT and MPV. While our GWAS did not find any new associations, our functional analyses revealed that genes in these regions influence thrombopoiesis and encode kinases, membrane proteins, proteins involved in cellular trafficking, transcription factors, proteasome complex subunits, proteins of signal transduction pathways, proteins involved in megakaryocyte development, and platelet production and hemostasis. PheWAS using a single-SNP Bonferroni correction for 1,368 diagnoses (0.05/1368 = 3.6E-5) revealed that several variants in these genes have pleiotropic associations with myocardial infarction, autoimmune, and hematologic disorders. We conclude that multiple genetic loci influence interindividual variation in platelet traits and also have significant pleiotropic effects; the related genes are in multiple functional pathways including those relevant to thrombopoiesis.
Collapse
Affiliation(s)
- Khader Shameer
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Joshua C. Denny
- Departments of Medicine and Biomedical Informatics, Vanderbilt University, Nashville, TN 37232, USA
| | - Keyue Ding
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Hayan Jouni
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - David R. Crosslin
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Mariza de Andrade
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Christopher G. Chute
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Peggy Peissig
- Biomedical Informatics Research Center, Marshfield Clinic, Marshfield, WI, 54449, USA
| | - Jennifer A. Pacheco
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rongling Li
- Office of Population Genomics, National Human Genome Research Institute, 5635 Fishers Lane, Suite 3058, MSC 9307, Bethesda, MD, 20892, USA
| | - Lisa Bastarache
- Departments of Medicine and Biomedical Informatics, Vanderbilt University, Nashville, TN 37232, USA
| | - Abel N. Kho
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Marylyn D Ritchie
- Center for Systems Genomics, Pennsylvania State University, Eberly College of Science, The Huck Institutes of the Life Sciences, 512 Wartik Laboratory, University Park, PA 16802 USA
| | - Daniel R. Masys
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Room 416 Eskind Medical Library, Nashville, TN, 37232, USA
| | - Rex L. Chisholm
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Eric B. Larson
- Group Health Research Institute, 1730 Minor Avenue, Suite 1600, Seattle, WA, 98101, USA
| | | | - Dan M. Roden
- Department of Pharmacology, Vanderbilt University School of Medicine, 1285 Medical Research Building IV, Nashville, TN, 37232, USA
| | - Gail P. Jarvik
- Department of Genome Sciences, University of Washington, 3720 15th Ave NE, Seattle WA 98195, USA
| | - Iftikhar J. Kullo
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
104
|
Weber FD, Wiesinger C, Forss-Petter S, Regelsberger G, Einwich A, Weber WHA, Köhler W, Stockinger H, Berger J. X-linked adrenoleukodystrophy: very long-chain fatty acid metabolism is severely impaired in monocytes but not in lymphocytes. Hum Mol Genet 2013; 23:2542-50. [PMID: 24363066 PMCID: PMC3990157 DOI: 10.1093/hmg/ddt645] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a fatal neurodegenerative disease caused by mutations in the ABCD1 gene, encoding a member of the peroxisomal ABC transporter family. The ABCD1 protein transports CoA-activated very long-chain fatty acids (VLCFAs) into peroxisomes for degradation via β-oxidation. In the severest form, X-ALD patients suffer from inflammatory demyelination of the brain. As the extent of the metabolic defect in the main immune cells is unknown, we explored their phenotypes concerning mRNA expression pattern of the three peroxisomal ABC transporters, VLCFA accumulation and peroxisomal β-oxidation. In controls, ABCD1 expression was high in monocytes, intermediate in B cells and low in T cells; ABCD2 expression was extremely low in monocytes, intermediate in B cells and highest in T cells; ABCD3 mRNA was equally distributed. In X-ALD patients, the expression patterns remained unaltered; accordingly, monocytes, which lack compensatory VLCFA transport by ABCD2, displayed the severest biochemical phenotype with a 6-fold accumulation of C26:0 and a striking 70% reduction in peroxisomal β-oxidation activity. In contrast, VLCFA metabolism was close to control values in B cells and T cells, supporting the hypothesis that sufficient ABCD2 is present to compensate for ABCD1 deficiency. Thus, the vulnerability of the main immune cell types is highly variable in X-ALD. Based on these results, we propose that in X-ALD the halt of inflammation after allogeneic hematopoietic stem cell transplantation relies particularly on the replacement of the monocyte lineage. Additionally, these findings support the concept that ABCD2 is a target for pharmacological induction as an alternative therapeutic strategy.
Collapse
Affiliation(s)
- Franziska D Weber
- Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna A-1090, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Chacon D, Beck D, Perera D, Wong JWH, Pimanda JE. BloodChIP: a database of comparative genome-wide transcription factor binding profiles in human blood cells. Nucleic Acids Res 2013; 42:D172-7. [PMID: 24185696 PMCID: PMC3964976 DOI: 10.1093/nar/gkt1036] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The BloodChIP database (http://www.med.unsw.edu.au/CRCWeb.nsf/page/BloodChIP) supports exploration and visualization of combinatorial transcription factor (TF) binding at a particular locus in human CD34-positive and other normal and leukaemic cells or retrieval of target gene sets for user-defined combinations of TFs across one or more cell types. Increasing numbers of genome-wide TF binding profiles are being added to public repositories, and this trend is likely to continue. For the power of these data sets to be fully harnessed by experimental scientists, there is a need for these data to be placed in context and easily accessible for downstream applications. To this end, we have built a user-friendly database that has at its core the genome-wide binding profiles of seven key haematopoietic TFs in human stem/progenitor cells. These binding profiles are compared with binding profiles in normal differentiated and leukaemic cells. We have integrated these TF binding profiles with chromatin marks and expression data in normal and leukaemic cell fractions. All queries can be exported into external sites to construct TF–gene and protein–protein networks and to evaluate the association of genes with cellular processes and tissue expression.
Collapse
Affiliation(s)
- Diego Chacon
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | | | | | | | | |
Collapse
|
106
|
Banerji CRS, Miranda-Saavedra D, Severini S, Widschwendter M, Enver T, Zhou JX, Teschendorff AE. Cellular network entropy as the energy potential in Waddington's differentiation landscape. Sci Rep 2013; 3:3039. [PMID: 24154593 PMCID: PMC3807110 DOI: 10.1038/srep03039] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 10/08/2013] [Indexed: 02/08/2023] Open
Abstract
Differentiation is a key cellular process in normal tissue development that is significantly altered in cancer. Although molecular signatures characterising pluripotency and multipotency exist, there is, as yet, no single quantitative mark of a cellular sample's position in the global differentiation hierarchy. Here we adopt a systems view and consider the sample's network entropy, a measure of signaling pathway promiscuity, computable from a sample's genome-wide expression profile. We demonstrate that network entropy provides a quantitative, in-silico, readout of the average undifferentiated state of the profiled cells, recapitulating the known hierarchy of pluripotent, multipotent and differentiated cell types. Network entropy further exhibits dynamic changes in time course differentiation data, and in line with a sample's differentiation stage. In disease, network entropy predicts a higher level of cellular plasticity in cancer stem cell populations compared to ordinary cancer cells. Importantly, network entropy also allows identification of key differentiation pathways. Our results are consistent with the view that pluripotency is a statistical property defined at the cellular population level, correlating with intra-sample heterogeneity, and driven by the degree of signaling promiscuity in cells. In summary, network entropy provides a quantitative measure of a cell's undifferentiated state, defining its elevation in Waddington's landscape.
Collapse
Affiliation(s)
- Christopher R. S. Banerji
- Statistical Cancer Genomics, Paul O'Gorman Building, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, United Kingdom
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London WC1E6BT United Kingdom
| | - Diego Miranda-Saavedra
- Bioinformatics and Genomics Laboratory, World Premier International (WPI) Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Simone Severini
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London WC1E6BT United Kingdom
- Department of Computer Science, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Martin Widschwendter
- Department of Women's Cancer, University College London, London WC1E 6BT, United Kingdom
| | - Tariq Enver
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, United Kingdom
| | - Joseph X. Zhou
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109-5234, USA
| | - Andrew E. Teschendorff
- Statistical Cancer Genomics, Paul O'Gorman Building, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, United Kingdom
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London WC1E6BT United Kingdom
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai Institute for Biological Sciences, 320 Yue Yang Road, Shanghai 200031, China
| |
Collapse
|
107
|
Diez D, Hutchins AP, Miranda-Saavedra D. Systematic identification of transcriptional regulatory modules from protein-protein interaction networks. Nucleic Acids Res 2013; 42:e6. [PMID: 24137002 PMCID: PMC3874207 DOI: 10.1093/nar/gkt913] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transcription factors (TFs) combine with co-factors to form transcriptional regulatory modules (TRMs) that regulate gene expression programs with spatiotemporal specificity. Here we present a novel and generic method (rTRM) for the reconstruction of TRMs that integrates genomic information from TF binding, cell type-specific gene expression and protein–protein interactions. rTRM was applied to reconstruct the TRMs specific for embryonic stem cells (ESC) and hematopoietic stem cells (HSC), neural progenitor cells, trophoblast stem cells and distinct types of terminally differentiated CD4+ T cells. The ESC and HSC TRM predictions were highly precise, yielding 77 and 96 proteins, of which ∼75% have been independently shown to be involved in the regulation of these cell types. Furthermore, rTRM successfully identified a large number of bridging proteins with known roles in ESCs and HSCs, which could not have been identified using genomic approaches alone, as they lack the ability to bind specific DNA sequences. This highlights the advantage of rTRM over other methods that ignore PPI information, as proteins need to interact with other proteins to form complexes and perform specific functions. The prediction and experimental validation of the co-factors that endow master regulatory TFs with the capacity to select specific genomic sites, modulate the local epigenetic profile and integrate multiple signals will provide important mechanistic insights not only into how such TFs operate, but also into abnormal transcriptional states leading to disease.
Collapse
Affiliation(s)
- Diego Diez
- World Premier International (WPI) Immunology Frontier Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita 565-0871, Osaka, Japan, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Ave, Guangzhou 510663, China and Fibrosis Laboratories, Institute of Cellular Medicine, Newcastle University Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | | | | |
Collapse
|
108
|
Xiang P, Wei W, Lo C, Rosten P, Hou J, Hoodless PA, Bilenky M, Bonifer C, Cockerill PN, Kirkpatrick A, Gottgens B, Hirst M, Humphries KR. Delineating MEIS1 cis-regulatory elements active in hematopoietic cells. Leukemia 2013; 28:433-6. [PMID: 24097337 DOI: 10.1038/leu.2013.287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- P Xiang
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - W Wei
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - C Lo
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - P Rosten
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - J Hou
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - P A Hoodless
- 1] Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada [2] University of British Columbia, Medical Genetics, Vancouver, British Columbia, Canada
| | - M Bilenky
- BC Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - C Bonifer
- School of Cancer Sciences, College of Medical and Dental Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| | - P N Cockerill
- School of Immunity and Infection, College of Medical and Dental Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| | - A Kirkpatrick
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - B Gottgens
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - M Hirst
- BC Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - K R Humphries
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| |
Collapse
|
109
|
Senis YA. Protein-tyrosine phosphatases: a new frontier in platelet signal transduction. J Thromb Haemost 2013; 11:1800-13. [PMID: 24015866 DOI: 10.1111/jth.12359] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Indexed: 08/31/2023]
Abstract
Platelet activation must be tightly controlled in order to allow platelets to respond rapidly to vascular injury and prevent thrombosis from occurring. Protein-tyrosine phosphorylation is one of the main ways in which activation signals are transmitted in platelets. Although much is known about the protein-tyrosine kinases (PTKs) that initiate and propagate activation signals, relatively little is known about the protein-tyrosine phosphatases (PTPs) that modulate these signals in platelets. PTPs are a family of enzymes that dephosphorylate tyrosine residues in proteins and regulate signals transmitted within cells. PTPs have been implicated in a variety of pathological conditions, including cancer, diabetes and autoimmunity, but their functions in hemostasis and thrombosis remain largely undefined. Exciting new findings from a number of groups have revealed that PTPs are in fact critical regulators of platelet activation and thrombosis. The primary aim of this review is to highlight the unique and important functions of PTPs in regulating platelet activity. Establishing the functions of PTPs in platelets is essential to better understand the molecular basis of thrombosis and may lead to the development of improved antithrombotic therapies.
Collapse
Affiliation(s)
- Y A Senis
- Centre for Cardiovascular and Respiratory Sciences, Institute of Biomedical Research, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| |
Collapse
|
110
|
Genome-wide analysis of transcriptional regulators in human HSPCs reveals a densely interconnected network of coding and noncoding genes. Blood 2013; 122:e12-22. [PMID: 23974199 DOI: 10.1182/blood-2013-03-490425] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Genome-wide combinatorial binding patterns for key transcription factors (TFs) have not been reported for primary human hematopoietic stem and progenitor cells (HSPCs), and have constrained analysis of the global architecture of molecular circuits controlling these cells. Here we provide high-resolution genome-wide binding maps for a heptad of key TFs (FLI1, ERG, GATA2, RUNX1, SCL, LYL1, and LMO2) in human CD34(+) HSPCs, together with quantitative RNA and microRNA expression profiles. We catalog binding of TFs at coding genes and microRNA promoters, and report that combinatorial binding of all 7 TFs is favored and associated with differential expression of genes and microRNA in HSPCs. We also uncover a previously unrecognized association between FLI1 and RUNX1 pairing in HSPCs, we establish a correlation between the density of histone modifications that mark active enhancers and the number of overlapping TFs at a peak, we demonstrate bivalent histone marks at promoters of heptad target genes in CD34(+) cells that are poised for later expression, and we identify complex relationships between specific microRNAs and coding genes regulated by the heptad. Taken together, these data reveal the power of integrating multifactor sequencing of chromatin immunoprecipitates with coding and noncoding gene expression to identify regulatory circuits controlling cell identity.
Collapse
|
111
|
McHale CM, Zhang L, Thomas R, Smith MT. Analysis of the transcriptome in molecular epidemiology studies. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2013; 54:500-517. [PMID: 23907930 PMCID: PMC5142298 DOI: 10.1002/em.21798] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 06/07/2013] [Accepted: 06/08/2013] [Indexed: 05/29/2023]
Abstract
The human transcriptome is complex, comprising multiple transcript types, mostly in the form of non-coding RNA (ncRNA). The majority of ncRNA is of the long form (lncRNA, ≥ 200 bp), which plays an important role in gene regulation through multiple mechanisms including epigenetics, chromatin modification, control of transcription factor binding, and regulation of alternative splicing. Both mRNA and ncRNA exhibit additional variability in the form of alternative splicing and RNA editing. All aspects of the human transcriptome can potentially be dysregulated by environmental exposures. Next-generation RNA sequencing (RNA-Seq) is the best available methodology to measure this although it has limitations, including experimental bias. The third phase of the MicroArray Quality Control Consortium project (MAQC-III), also called Sequencing Quality Control (SeQC), aims to address these limitations through standardization of experimental and bioinformatic methodologies. A limited number of toxicogenomic studies have been conducted to date using RNA-Seq. This review describes the complexity of the human transcriptome, the application of transcriptomics by RNA-Seq or microarray in molecular epidemiology studies, and limitations of these approaches including the type of cell or tissue analyzed, experimental variation, and confounding. By using good study designs with precise, individual exposure measurements, sufficient power and incorporation of phenotypic anchors, studies in human populations can identify biomarkers of exposure and/or early effect and elucidate mechanisms of action underlying associated diseases, even at low doses. Analysis of datasets at the pathway level can compensate for some of the limitations of RNA-Seq and, as more datasets become available, will increasingly elucidate the exposure-disease continuum.
Collapse
Affiliation(s)
- Cliona M McHale
- Division of Environmental Health Sciences, Genes and Environment Laboratory, School of Public Health, University of California, Berkeley, California 94720, USA.
| | | | | | | |
Collapse
|
112
|
Gaujoux R, Seoighe C. CellMix: a comprehensive toolbox for gene expression deconvolution. Bioinformatics 2013; 29:2211-2. [DOI: 10.1093/bioinformatics/btt351] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
113
|
Interferon-beta therapy in multiple sclerosis: the short-term and long-term effects on the patients' individual gene expression in peripheral blood. Mol Neurobiol 2013; 48:737-56. [PMID: 23636981 DOI: 10.1007/s12035-013-8463-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/16/2013] [Indexed: 01/17/2023]
Abstract
Therapy with interferon-beta (IFN-beta) is a mainstay in the management of relapsing-remitting multiple sclerosis (MS), with proven long-term effectiveness and safety. Much has been learned about the molecular mechanisms of action of IFN-beta in the past years. Previous studies described more than a hundred genes to be modulated in expression in blood cells in response to the therapy. However, for many of these genes, the precise temporal expression pattern and the therapeutic relevance are unclear. We used Affymetrix microarrays to investigate in more detail the gene expression changes in peripheral blood mononuclear cells from MS patients receiving subcutaneous IFN-beta-1a. The blood samples were obtained longitudinally at five different time points up to 2 years after the start of therapy, and the patients were clinically followed up for 5 years. We examined the functions of the genes that were upregulated or downregulated at the transcript level after short-term or long-term treatment. Moreover, we analyzed their mutual interactions and their regulation by transcription factors. Compared to pretreatment levels, 96 genes were identified as highly differentially expressed, many of them already after the first IFN-beta injection. The interactions between these genes form a large network with multiple feedback loops, indicating the complex crosstalk between innate and adaptive immune responses during therapy. We discuss the genes and biological processes that might be important to reduce disease activity by attenuating the proliferation of autoreactive immune cells and their migration into the central nervous system. In summary, we present novel insights that extend the current knowledge on the early and late pharmacodynamic effects of IFN-beta therapy and describe gene expression differences between the individual patients that reflect clinical heterogeneity.
Collapse
|
114
|
Cvejic A, Haer-Wigman L, Stephens JC, Kostadima M, Smethurst PA, Frontini M, van den Akker E, Bertone P, Bielczyk-Maczyńska E, Farrow S, Fehrmann RSN, Gray A, de Haas M, Haver VG, Jordan G, Karjalainen J, Kerstens HHD, Kiddle G, Lloyd-Jones H, Needs M, Poole J, Soussan AA, Rendon A, Rieneck K, Sambrook JG, Schepers H, Silljé HHW, Sipos B, Swinkels D, Tamuri AU, Verweij N, Watkins NA, Westra HJ, Stemple D, Franke L, Soranzo N, Stunnenberg HG, Goldman N, van der Harst P, van der Schoot CE, Ouwehand WH, Albers CA. SMIM1 underlies the Vel blood group and influences red blood cell traits. Nat Genet 2013; 45:542-545. [PMID: 23563608 PMCID: PMC4179282 DOI: 10.1038/ng.2603] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 03/08/2013] [Indexed: 11/08/2022]
Abstract
The blood group Vel was discovered 60 years ago, but the underlying gene is unknown. Individuals negative for the Vel antigen are rare and are required for the safe transfusion of patients with antibodies to Vel. To identify the responsible gene, we sequenced the exomes of five individuals negative for the Vel antigen and found that four were homozygous and one was heterozygous for a low-frequency 17-nucleotide frameshift deletion in the gene encoding the 78-amino-acid transmembrane protein SMIM1. A follow-up study showing that 59 of 64 Vel-negative individuals were homozygous for the same deletion and expression of the Vel antigen on SMIM1-transfected cells confirm SMIM1 as the gene underlying the Vel blood group. An expression quantitative trait locus (eQTL), the common SNP rs1175550 contributes to variable expression of the Vel antigen (P = 0.003) and influences the mean hemoglobin concentration of red blood cells (RBCs; P = 8.6 × 10(-15)). In vivo, zebrafish with smim1 knockdown showed a mild reduction in the number of RBCs, identifying SMIM1 as a new regulator of RBC formation. Our findings are of immediate relevance, as the homozygous presence of the deletion allows the unequivocal identification of Vel-negative blood donors.
Collapse
Affiliation(s)
- Ana Cvejic
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1HH, United Kingdom
| | - Lonneke Haer-Wigman
- Department of Experimental Immunohaematology, Sanquin Research, 1066 CX, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1066 CX, The Netherlands
| | - Jonathan C Stephens
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
| | - Myrto Kostadima
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Peter A Smethurst
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
| | - Emile van den Akker
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1066 CX, The Netherlands
- Department of Hematopoiesis, Sanquin Research, Amsterdam, 1066 CX, The Netherlands
| | - Paul Bertone
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Ewa Bielczyk-Maczyńska
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1HH, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
| | - Samantha Farrow
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
| | - Rudolf SN Fehrmann
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, 9700 RB, The Netherlands
| | - Alan Gray
- NHS Blood and Transplant, Tooting, London, SW17 0RB, United Kingdom
| | - Masja de Haas
- Department of Experimental Immunohaematology, Sanquin Research, 1066 CX, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1066 CX, The Netherlands
| | - Vincent G Haver
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, 9700 RB, The Netherlands
| | | | - Juha Karjalainen
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, 9700 RB, The Netherlands
| | - Hindrik HD Kerstens
- Department of Molecular Biology, Faculty of Science, Nijmegen Centre for Molecular Life Sciences, Radboud University, Nijmegen, 6525 GA, The Netherlands
| | - Graham Kiddle
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
| | - Heather Lloyd-Jones
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
| | - Malcolm Needs
- NHS Blood and Transplant, Tooting, London, SW17 0RB, United Kingdom
| | - Joyce Poole
- International Blood Group Reference Laboratory, NHS Blood and Transplant, North Bristol Park, Northway, Filton, Bristol, BS34 7QH, United Kingdom
| | - Aicha Ait Soussan
- Department of Experimental Immunohaematology, Sanquin Research, 1066 CX, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1066 CX, The Netherlands
| | - Augusto Rendon
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
- MRC Biostatistics Unit, Institute of Public Health, Cambridge, CB2 0SR, United Kingdom
| | - Klaus Rieneck
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, Copenhagen, DK-2100, Denmark
| | - Jennifer G Sambrook
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
| | - Hein Schepers
- University of Groningen, University Medical Center Groningen, Department of Experimental Hematology, Groningen, 9700 RB, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Stem Cell Biology, Groningen, 9700 RB, The Netherlands
| | - Herman H W Silljé
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, 9700 RB, The Netherlands
| | - Botond Sipos
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Dorine Swinkels
- Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic diseases, Radboud University Medical Centre, Nijmegen, 6500 HB, The Netherlands
| | - Asif U Tamuri
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Niek Verweij
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, 9700 RB, The Netherlands
| | | | - Harm-Jan Westra
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, 9700 RB, The Netherlands
| | - Derek Stemple
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1HH, United Kingdom
| | - Lude Franke
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, 9700 RB, The Netherlands
| | - Nicole Soranzo
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1HH, United Kingdom
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculty of Science, Nijmegen Centre for Molecular Life Sciences, Radboud University, Nijmegen, 6525 GA, The Netherlands
| | - Nick Goldman
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, 9700 RB, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, 9700 RB, The Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohaematology, Sanquin Research, 1066 CX, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1066 CX, The Netherlands
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1HH, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
| | - Cornelis A Albers
- Department of Haematology, University of Cambridge, CB2 0PT, United Kingdom
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1HH, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, United Kingdom
- NHS Blood and Transplant, Cambridge, CB2 0PT, United Kingdom
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, 6500 HB, The Netherlands
| |
Collapse
|
115
|
Goggs R, Savage JS, Mellor H, Poole AW. The small GTPase Rif is dispensable for platelet filopodia generation in mice. PLoS One 2013; 8:e54663. [PMID: 23359340 PMCID: PMC3554654 DOI: 10.1371/journal.pone.0054663] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 12/13/2012] [Indexed: 11/18/2022] Open
Abstract
Background Formation of filopodia and other shape change events are vital for platelet hemostatic function. The mechanisms regulating filopodia formation by platelets are incompletely understood however. In particular the small GTPase responsible for initiating filopodia formation by platelets remains elusive. The canonical pathway involving Cdc42 is not essential for filopodia formation in mouse platelets. The small GTPase Rif (RhoF) provides an alternative route to filopodia generation in other cell types and is expressed in both human and mouse platelets. Hypothesis/Objective We hypothesized that Rif might be responsible for generating filopodia by platelets and generated a novel knockout mouse model to investigate the functional role of Rif in platelets. Methodology/Principal Findings Constitutive RhoF−/− mice are viable and have normal platelet, leukocyte and erythrocyte counts and indices. RhoF−/− platelets form filopodia and spread normally on various agonist surfaces in static conditions and under arterial shear. In addition, RhoF−/− platelets have normal actin dynamics, are able to activate and aggregate normally and secrete from alpha and dense granules in response to collagen related peptide and thrombin stimulation. Conclusions The small GTPase Rif does not appear to be critical for platelet function in mice. Functional overlap between Rif and other small GTPases may be responsible for the non-essential role of Rif in platelets.
Collapse
Affiliation(s)
- Robert Goggs
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
| | - Joshua S. Savage
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
| | - Harry Mellor
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Alastair W. Poole
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
116
|
Göttgens B. Genome-scale technology driven advances to research into normal and malignant haematopoiesis. SCIENTIFICA 2012; 2012:437956. [PMID: 24278696 PMCID: PMC3820533 DOI: 10.6064/2012/437956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 12/16/2012] [Indexed: 06/02/2023]
Abstract
Haematopoiesis or blood development has long served as a model system for adult stem cell biology. Moreover, when combined, the various cancers of the blood represent one of the commonest human malignancies. Large numbers of researchers have therefore dedicated their scientific careers to studying haematopoiesis for more than a century. Throughout this period, many new technologies have first been applied towards the study of blood cells, and the research fields of normal and malignant haematopoiesis have also been some of the earliest adopters of genome-scale technologies. This has resulted in significant new insights with implications ranging from basic biological mechanisms to patient diagnosis and prognosis and also produced lessons likely to be relevant for many other areas of biomedical research. This paper discusses the current state of play for a range of genome-scale applications within haemopoiesis research, including gene expression profiling, ChIP-sequencing, genomewide association analysis, and cancer genome sequencing. A concluding outlook section explores likely future areas of progress as well as potential technological and educational bottlenecks.
Collapse
Affiliation(s)
- Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge University and Wellcome Trust and MRC Stem Cell Institute, Hills Road, Cambridge CB2 0XY, UK
| |
Collapse
|
117
|
A GWAS sequence variant for platelet volume marks an alternative DNM3 promoter in megakaryocytes near a MEIS1 binding site. Blood 2012; 120:4859-68. [PMID: 22972982 PMCID: PMC3520622 DOI: 10.1182/blood-2012-01-401893] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
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.
Collapse
|
118
|
Abstract
PURPOSE OF REVIEW This review summarizes our current knowledge of common gene variants (polymorphisms) that have small individual effects on platelet function in humans, but can cumulatively lead to hyperreactive platelets and increase risk for negative outcomes in thrombotic disorders. RECENT FINDINGS Candidate gene association and genome-wide association studies (GWAS) have identified loci that include single nucleotide polymorphisms, which exert a cumulative effect on platelet function by modifying basic platelet parameters, such as mean platelet volume (MPV) or platelet count, by altering the expression or activity of key platelet receptors, or by influencing downstream effector pathways utilized by these receptors. SUMMARY Variation in MPV between normal individuals is responsible for roughly a two-fold range in platelet protein content, including key surface receptors and reactive granule constituents, the association of ADRA2, GP1BA, GP6, ITGA2 and P2Y12 variants with platelet reactivity, initially identified by candidate gene analyses, has now been validated by genome-wide approaches in much larger individual cohorts, and GWAS have identified novel gene variants, most notably PEAR1, that participate in variation in platelet reactivity among normal individuals, all of which contribute to a genetic basis for differences in platelet reactivty among normal individuals.
Collapse
|
119
|
Abstract
Wnt signaling is involved in numerous aspects of vertebrate development and homeostasis, including the formation and function of blood cells. Here, we show that canonical and noncanonical Wnt signaling pathways are present and functional in megakaryocytes (MKs), with several Wnt effectors displaying MK-restricted expression. Using the CHRF288-11 cell line as a model for human MKs, the canonical Wnt3a signal was found to induce a time and dose-dependent increase in β-catenin expression. β-catenin accumulation was inhibited by the canonical antagonist dickkopf-1 (DKK1) and by the noncanonical agonist Wnt5a. Whole genome expression analysis demonstrated that Wnt3a and Wnt5a regulated distinct patterns of gene expression in MKs, and revealed a further interplay between canonical and noncanonical Wnt pathways. Fetal liver cells derived from low-density-lipoprotein receptor-related protein 6-deficient mice (LRP6(-/-)), generated dramatically reduced numbers of MKs in culture of lower ploidy (2N and 4N) than wild-type controls, implicating LRP6-dependent Wnt signaling in MK proliferation and maturation. Finally, in wild-type mature murine fetal liver-derived MKs, Wnt3a potently induced proplatelet formation, an effect that could be completely abrogated by DKK1. These data identify novel extrinsic regulators of proplatelet formation, and reveal a profound role for Wnt signaling in platelet production.
Collapse
|
120
|
Jupe S, Akkerman JW, Soranzo N, Ouwehand WH. Reactome - a curated knowledgebase of biological pathways: megakaryocytes and platelets. J Thromb Haemost 2012; 10:2399-402. [PMID: 22985186 PMCID: PMC3578965 DOI: 10.1111/j.1538-7836.2012.04930.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The platelet field is undergoing a radical transformation from reductionist simplification to large scale integration. Following the era of simplification whereby biological processes were dissected at the molecular and atomic level, new technologies have now generated an overwhelming flow of information that can only be comprehended in an integrated approach. High throughput analyses of transcription and translation in megakaryocytes and platelets, individual analyses of membranes and secretory granules, the clustering of pathways for platelet activation and inhibition in signalosomes all add to a complexity that requires platforms for knowledge accumulation. Here we introduce Reactome, a curated knowledgebase of biological pathways with extensive coverage of pathways relevant to megakaryocytes, platelets and haemostasis. This resource is compared with other data resources for platelets, e.g. the Platelet Web.
Collapse
Affiliation(s)
- S Jupe
- European Bioinformatics Institute, Hinxton, Cambridge, UK Department of Clinical Chemistry and Haematology, University Medical Centre Utrecht, Utrecht, the Netherlands The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge Department of Haematology, University of Cambridge and National Health Service Blood and Transplant, Cambridge, UK
| | | | | | | |
Collapse
|
121
|
Systematic analysis of microRNA fingerprints in thrombocythemic platelets using integrated platforms. Blood 2012; 120:3575-85. [PMID: 22869791 DOI: 10.1182/blood-2012-02-411264] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Posttranscriptional and translational controls mediated by microRNAs (miRNA) regulate diverse biologic processes. We dissected regulatory effects of miRNAs relevant to megakaryocytopoiesis and platelet biology by analyzing expression patterns in 79 subjects with thrombocytosis and controls, and integrated data with transcriptomic and proteomic platforms. We validated a unique 21-miRNA genetic fingerprint associated with thrombocytosis, and demonstrated that a 3-member subset defines essential thrombocythemia (ET). The genetic signature includes functional guide and passenger strands of the previously uncharacterized miR 490 (5p and 3p), which displayed restricted, low-level expression in megakaryocytes/platelets (compared with leukocytes), and aberrant expression during thrombocytosis, most profound in ET. Overexpression of miR 490 in a bilineage differentiation model of megakaryocyte/erythroid progenitor formation was insufficient for hematopoietic colony differentiation and/or lineage specification. Integration of transcriptomic and mass spectrometric datasets with functional reporter assays identified dishevelled associated activator of morphogenesis 1 (DAAM1) as a miR 490 5p protein target demonstrating decreased expression in ET platelets, putatively by translational control (and not by mRNA target degradation). Our data define a dysregulated miRNA fingerprint in thrombocytosis and support a developmentally restricted function of miR 490 (and its putative DAAM1 target) to conditions associated with exaggerated megakaryocytopoiesis and/or proplatelet formation.
Collapse
|
122
|
A Meta-Analysis of Gene Expression Data Identifies a Molecular Signature Characteristic for Tumor-Stage Mycosis Fungoides. J Invest Dermatol 2012; 132:2050-9. [DOI: 10.1038/jid.2012.117] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
123
|
Drew JE. Cellular defense system gene expression profiling of human whole blood: opportunities to predict health benefits in response to diet. Adv Nutr 2012; 3:499-505. [PMID: 22797985 PMCID: PMC3649718 DOI: 10.3945/an.112.002121] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Diet is a critical factor in the maintenance of human cellular defense systems, immunity, inflammation, redox regulation, metabolism, and DNA repair that ensure optimal health and reduce disease risk. Assessment of dietary modulation of cellular defense systems in humans has been limited due to difficulties in accessing target tissues. Notably, peripheral blood gene expression profiles associated with nonhematologic disease are detectable. Coupled with recent innovations in gene expression technologies, gene expression profiling of human blood to determine predictive markers associated with health status and dietary modulation is now a feasible prospect for nutrition scientists. This review focuses on cellular defense system gene expression profiling of human whole blood and the opportunities this presents, using recent technological advances, to predict health status and benefits conferred by diet.
Collapse
|
124
|
Londoño MC, Danger R, Giral M, Soulillou JP, Sánchez-Fueyo A, Brouard S. A need for biomarkers of operational tolerance in liver and kidney transplantation. Am J Transplant 2012; 12:1370-7. [PMID: 22486792 DOI: 10.1111/j.1600-6143.2012.04035.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Both kidney and particularly liver recipients can occasionally discontinue all immunosuppressive drugs without undergoing rejection. These patients, who maintain stable graft function off immunosuppressive drugs without clinically significant detrimental immune responses and/or immune deficits, are conventionally termed operationally tolerant and offer a unique paradigm of tolerance in humans. The immune characterization of operationally tolerant transplant recipients has recently received substantial attention. Operationally tolerant patients might exhibit a signature of tolerance that could potentially be useful to select recipients amenable to drug minimization or withdrawal. Furthermore, elucidation of the molecular pathways associated with the operational tolerance phenotype could provide novel targets for therapy. Particular emphasis has been placed on the use of blood samples and high-throughput transcriptional profiling techniques. In liver transplantation, natural killer related transcripts seem to be the most robust markers of operational tolerance, whereas enrichment in B cell-related gene expression markers has been consistently found in blood samples from operationally tolerant kidney recipients, suggesting that different mechanisms operate in the two situations. In this minireview, we summarize the main achievements of recently published reports focused on the identification of transcriptional markers of operational tolerance, we highlight their mechanistic and clinical implications and describe their methodological limitations.
Collapse
Affiliation(s)
- M-C Londoño
- Liver Transplant Unit, Hospital Clinic, IDIBAPS, CIBEREHD, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
125
|
Vaiyapuri S, Jones CI, Sasikumar P, Moraes LA, Munger SJ, Wright JR, Ali MS, Sage T, Kaiser WJ, Tucker KL, Stain CJ, Bye AP, Jones S, Oviedo-Orta E, Simon AM, Mahaut-Smith MP, Gibbins JM. Gap junctions and connexin hemichannels underpin hemostasis and thrombosis. Circulation 2012; 125:2479-91. [PMID: 22528526 PMCID: PMC3378664 DOI: 10.1161/circulationaha.112.101246] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Connexins are a widespread family of membrane proteins that assemble into hexameric hemichannels, also known as connexons. Connexons regulate membrane permeability in individual cells or couple between adjacent cells to form gap junctions and thereby provide a pathway for regulated intercellular communication. We have examined the role of connexins in platelets, blood cells that circulate in isolation but on tissue injury adhere to each other and the vessel wall to prevent blood loss and to facilitate wound repair. METHODS AND RESULTS We report the presence of connexins in platelets, notably connexin37, and that the formation of gap junctions within platelet thrombi is required for the control of clot retraction. Inhibition of connexin function modulated a range of platelet functional responses before platelet-platelet contact and reduced laser-induced thrombosis in vivo in mice. Deletion of the Cx37 gene (Gja4) in transgenic mice reduced platelet aggregation, fibrinogen binding, granule secretion, and clot retraction, indicating an important role for connexin37 hemichannels and gap junctions in platelet thrombus function. CONCLUSIONS Together, these data demonstrate that platelet gap junctions and hemichannels underpin the control of hemostasis and thrombosis and represent potential therapeutic targets.
Collapse
Affiliation(s)
- Sakthivel Vaiyapuri
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Chris I. Jones
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Parvathy Sasikumar
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Leonardo A. Moraes
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Joy R. Wright
- Dept of Cell Physiology & Pharmacology, University of Leicester, Leicester
| | - Marfoua S. Ali
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Tanya Sage
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - William J. Kaiser
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Katherine L. Tucker
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Alexander P. Bye
- Dept of Cell Physiology & Pharmacology, University of Leicester, Leicester
| | - Sarah Jones
- Dept of Cell Physiology & Pharmacology, University of Leicester, Leicester
| | - Ernesto Oviedo-Orta
- Cardiovascular Biology Research, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | | | | | - Jonathan M. Gibbins
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
126
|
Houseman EA, Accomando WP, Koestler DC, Christensen BC, Marsit CJ, Nelson HH, Wiencke JK, Kelsey KT. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics 2012; 13:86. [PMID: 22568884 PMCID: PMC3532182 DOI: 10.1186/1471-2105-13-86] [Citation(s) in RCA: 2362] [Impact Index Per Article: 181.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 04/20/2012] [Indexed: 12/14/2022] Open
Abstract
Background There has been a long-standing need in biomedical research for a method that quantifies the normally mixed composition of leukocytes beyond what is possible by simple histological or flow cytometric assessments. The latter is restricted by the labile nature of protein epitopes, requirements for cell processing, and timely cell analysis. In a diverse array of diseases and following numerous immune-toxic exposures, leukocyte composition will critically inform the underlying immuno-biology to most chronic medical conditions. Emerging research demonstrates that DNA methylation is responsible for cellular differentiation, and when measured in whole peripheral blood, serves to distinguish cancer cases from controls. Results Here we present a method, similar to regression calibration, for inferring changes in the distribution of white blood cells between different subpopulations (e.g. cases and controls) using DNA methylation signatures, in combination with a previously obtained external validation set consisting of signatures from purified leukocyte samples. We validate the fundamental idea in a cell mixture reconstruction experiment, then demonstrate our method on DNA methylation data sets from several studies, including data from a Head and Neck Squamous Cell Carcinoma (HNSCC) study and an ovarian cancer study. Our method produces results consistent with prior biological findings, thereby validating the approach. Conclusions Our method, in combination with an appropriate external validation set, promises new opportunities for large-scale immunological studies of both disease states and noxious exposures.
Collapse
Affiliation(s)
- Eugene Andres Houseman
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Yamashiro S, Gokhin DS, Kimura S, Nowak RB, Fowler VM. Tropomodulins: pointed-end capping proteins that regulate actin filament architecture in diverse cell types. Cytoskeleton (Hoboken) 2012; 69:337-70. [PMID: 22488942 DOI: 10.1002/cm.21031] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 03/23/2012] [Accepted: 03/26/2012] [Indexed: 01/31/2023]
Abstract
Tropomodulins are a family of four proteins (Tmods 1-4) that cap the pointed ends of actin filaments in actin cytoskeletal structures in a developmentally regulated and tissue-specific manner. Unique among capping proteins, Tmods also bind tropomyosins (TMs), which greatly enhance the actin filament pointed-end capping activity of Tmods. Tmods are defined by a TM-regulated/Pointed-End Actin Capping (TM-Cap) domain in their unstructured N-terminal portion, followed by a compact, folded Leucine-Rich Repeat/Pointed-End Actin Capping (LRR-Cap) domain. By inhibiting actin monomer association and dissociation from pointed ends, Tmods regulate actin dynamics and turnover, stabilizing actin filament lengths and cytoskeletal architecture. In this review, we summarize the genes, structural features, molecular and biochemical properties, actin regulatory mechanisms, expression patterns, and cell and tissue functions of Tmods. By understanding Tmods' functions in the context of their molecular structure, actin regulation, binding partners, and related variants (leiomodins 1-3), we can draw broad conclusions that can explain the diverse morphological and functional phenotypes that arise from Tmod perturbation experiments in vitro and in vivo. Tmod-based stabilization and organization of intracellular actin filament networks provide key insights into how the emergent properties of the actin cytoskeleton drive tissue morphogenesis and physiology.
Collapse
Affiliation(s)
- Sawako Yamashiro
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
128
|
Edelstein LC, Luna EJ, Gibson IB, Bray M, Jin Y, Kondkar A, Nagalla S, Hadjout-Rabi N, Smith TC, Covarrubias D, Jones SN, Ahmad F, Stolla M, Kong X, Fang Z, Bergmeier W, Shaw C, Leal SM, Bray PF. Human genome-wide association and mouse knockout approaches identify platelet supervillin as an inhibitor of thrombus formation under shear stress. Circulation 2012; 125:2762-71. [PMID: 22550155 DOI: 10.1161/circulationaha.112.091462] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND High shear force critically regulates platelet adhesion and thrombus formation during ischemic vascular events. To identify genetic factors that influence platelet thrombus formation under high shear stress, we performed a genome-wide association study and confirmatory experiments in human and animal platelets. METHODS AND RESULTS Closure times in the shear-dependent platelet function analyzer (PFA)-100 were measured on healthy, nondiabetic European Americans (n=125) and blacks (n=116). A genome-wide association (P<5×10(-8)) was identified with 2 single-nucleotide polymorphisms within the SVIL gene (chromosome 10p11.23) in African Americans but not European Americans. Microarray analyses of human platelet RNA demonstrated the presence of SVIL isoform 1 (supervillin) but not muscle-specific isoforms 2 and 3 (archvillin, SmAV). SVIL mRNA levels were associated with SVIL genotypes (P≤0.02) and were inversely correlated with PFA-100 closure times (P<0.04) and platelet volume (P<0.02). Leukocyte-depleted platelets contained abundant levels of the ≈205-kDa supervillin polypeptide. To assess functionality, mice lacking platelet supervillin were generated and back-crossed onto a C57BL/6 background. Compared with controls, murine platelets lacking supervillin were larger by flow cytometry and confocal microscopy and exhibited enhanced platelet thrombus formation under high-shear but not low-shear conditions. CONCLUSIONS We show for the first time that (1) platelets contain supervillin; (2) platelet thrombus formation in the PFA-100 is associated with human SVIL variants and low SVIL expression; and (3) murine platelets lacking supervillin exhibit enhanced platelet thrombus formation at high shear stress. These data are consistent with an inhibitory role for supervillin in platelet adhesion and arterial thrombosis.
Collapse
Affiliation(s)
- Leonard C Edelstein
- Cardeza Foundation for Hematologic Research, Department of Medicine, Jefferson Medical College, Thomas Jefferson University, Curtis Building, Room 324, 1015 Walnut St, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Qayyum R, Snively BM, Ziv E, Nalls MA, Liu Y, Tang W, Yanek LR, Lange L, Evans MK, Ganesh S, Austin MA, Lettre G, Becker DM, Zonderman AB, Singleton AB, Harris TB, Mohler ER, Logsdon BA, Kooperberg C, Folsom AR, Wilson JG, Becker LC, Reiner AP. A meta-analysis and genome-wide association study of platelet count and mean platelet volume in african americans. PLoS Genet 2012; 8:e1002491. [PMID: 22423221 PMCID: PMC3299192 DOI: 10.1371/journal.pgen.1002491] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 12/05/2011] [Indexed: 12/24/2022] Open
Abstract
Several genetic variants associated with platelet count and mean platelet volume (MPV) were recently reported in people of European ancestry. In this meta-analysis of 7 genome-wide association studies (GWAS) enrolling African Americans, our aim was to identify novel genetic variants associated with platelet count and MPV. For all cohorts, GWAS analysis was performed using additive models after adjusting for age, sex, and population stratification. For both platelet phenotypes, meta-analyses were conducted using inverse-variance weighted fixed-effect models. Platelet aggregation assays in whole blood were performed in the participants of the GeneSTAR cohort. Genetic variants in ten independent regions were associated with platelet count (N = 16,388) with p<5×10(-8) of which 5 have not been associated with platelet count in previous GWAS. The novel genetic variants associated with platelet count were in the following regions (the most significant SNP, closest gene, and p-value): 6p22 (rs12526480, LRRC16A, p = 9.1×10(-9)), 7q11 (rs13236689, CD36, p = 2.8×10(-9)), 10q21 (rs7896518, JMJD1C, p = 2.3×10(-12)), 11q13 (rs477895, BAD, p = 4.9×10(-8)), and 20q13 (rs151361, SLMO2, p = 9.4×10(-9)). Three of these loci (10q21, 11q13, and 20q13) were replicated in European Americans (N = 14,909) and one (11q13) in Hispanic Americans (N = 3,462). For MPV (N = 4,531), genetic variants in 3 regions were significant at p<5×10(-8), two of which were also associated with platelet count. Previously reported regions that were also significant in this study were 6p21, 6q23, 7q22, 12q24, and 19p13 for platelet count and 7q22, 17q11, and 19p13 for MPV. The most significant SNP in 1 region was also associated with ADP-induced maximal platelet aggregation in whole blood (12q24). Thus through a meta-analysis of GWAS enrolling African Americans, we have identified 5 novel regions associated with platelet count of which 3 were replicated in other ethnic groups. In addition, we also found one region associated with platelet aggregation that may play a potential role in atherothrombosis.
Collapse
Affiliation(s)
- Rehan Qayyum
- GeneSTAR Research Program, Division of General
Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United
States of America
| | - Beverly M. Snively
- Department of Biostatistical Sciences, Wake
Forest School of Medicine, Winston-Salem, North Carolina, United States of
America
| | - Elad Ziv
- Department of Medicine, University of
California San Francisco, San Francisco, California, United States of
America
| | - Michael A. Nalls
- Laboratory of Neurogenetics, National
Institute on Aging, National Institutes of Health, Bethesda, Maryland, United
States of America
| | - Yongmei Liu
- Department of Epidemiology and Prevention,
Division of Public Health Sciences, Wake Forest University School of Medicine,
Winston-Salem, North Carolina, United States of America
| | - Weihong Tang
- Division of Epidemiology and Community Health,
University of Minnesota School of Public Health, Minneapolis, Minnesota, United
States of America
| | - Lisa R. Yanek
- GeneSTAR Research Program, Division of General
Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United
States of America
| | - Leslie Lange
- Department of Genetics, School of Medicine,
The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina,
United States of America
| | - Michele K. Evans
- Health Disparities Research Section, Clinical
Research Branch, National Institute on Aging, National Institutes of Health,
Baltimore, Maryland, United States of America
| | - Santhi Ganesh
- Division of Cardiology, University of Michigan
Health System, Ann Arbor, Michigan, United States of America
| | - Melissa A. Austin
- Department of Epidemiology, University of
Washington, Seattle, Washington, United States of America
- Division of Public Health Sciences, Fred
Hutchinson Cancer Research Center, Seattle, Washington, United States of
America
| | | | - Diane M. Becker
- GeneSTAR Research Program, Division of General
Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United
States of America
| | - Alan B. Zonderman
- Laboratory of Personality and Cognition,
National Institute on Aging, National Institutes of Health, Baltimore, Maryland,
United States of America
| | - Andrew B. Singleton
- Laboratory of Neurogenetics, National
Institute on Aging, National Institutes of Health, Bethesda, Maryland, United
States of America
| | - Tamara B. Harris
- Laboratory for Epidemiology, Demography, and
Biometry, National Institute on Aging, National Institutes of Health, Baltimore,
Maryland, United States of America
| | - Emile R. Mohler
- Department of Medicine, University of
Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of
America
| | - Benjamin A. Logsdon
- Program in Biostatistics and Biomathematics,
Division of Public Health Sciences, Fred Hutchinson Cancer Research Center,
Seattle, Washington, United States of America
| | - Charles Kooperberg
- Program in Biostatistics and Biomathematics,
Division of Public Health Sciences, Fred Hutchinson Cancer Research Center,
Seattle, Washington, United States of America
| | - Aaron R. Folsom
- Division of Epidemiology and Community Health,
University of Minnesota School of Public Health, Minneapolis, Minnesota, United
States of America
| | - James G. Wilson
- Department of Medicine, University of
Mississippi Medical Center, Jackson, Mississippi, United States of
America
| | - Lewis C. Becker
- GeneSTAR Research Program, Division of General
Internal Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United
States of America
| | - Alexander P. Reiner
- Department of Epidemiology, University of
Washington, Seattle, Washington, United States of America
| |
Collapse
|
130
|
Compound inheritance of a low-frequency regulatory SNP and a rare null mutation in exon-junction complex subunit RBM8A causes TAR syndrome. Nat Genet 2012; 44:435-9, S1-2. [PMID: 22366785 PMCID: PMC3428915 DOI: 10.1038/ng.1083] [Citation(s) in RCA: 300] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 12/21/2011] [Indexed: 02/06/2023]
Abstract
The exon-junction complex (EJC) performs essential RNA processing tasks. Here, we describe the first human disorder, thrombocytopenia with absent radii (TAR), caused by deficiency in one of the four EJC subunits. Compound inheritance of a rare null allele and one of two low-frequency SNPs in the regulatory regions of RBM8A, encoding the Y14 subunit of EJC, causes TAR. We found that this inheritance mechanism explained 53 of 55 cases (P < 5 × 10(-228)) of the rare congenital malformation syndrome. Of the 53 cases with this inheritance pattern, 51 carried a submicroscopic deletion of 1q21.1 that has previously been associated with TAR, and two carried a truncation or frameshift null mutation in RBM8A. We show that the two regulatory SNPs result in diminished RBM8A transcription in vitro and that Y14 expression is reduced in platelets from individuals with TAR. Our data implicate Y14 insufficiency and, presumably, an EJC defect as the cause of TAR syndrome.
Collapse
|
131
|
Sivapalaratnam S, Basart H, Watkins NA, Maiwald S, Rendon A, Krishnan U, Sondermeijer BM, Creemers EE, Pinto-Sietsma SJ, Hovingh K, Ouwehand WH, Kastelein JJP, Goodall AH, Trip MD. Monocyte gene expression signature of patients with early onset coronary artery disease. PLoS One 2012; 7:e32166. [PMID: 22363809 PMCID: PMC3283726 DOI: 10.1371/journal.pone.0032166] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/19/2012] [Indexed: 11/18/2022] Open
Abstract
The burden of cardiovascular disease (CVD) cannot be fully addressed by therapy targeting known pathophysiological pathways. Even with stringent control of all risk factors CVD events are only diminished by half. A number of additional pathways probably play a role in the development of CVD and might serve as novel therapeutic targets. Genome wide expression studies represent a powerful tool to identify such novel pathways. We compared the expression profiles in monocytes from twenty two young male patients with premature familial CAD with those from controls matched for age, sex and smoking status, without a family history of CVD. Since all patients were on statins and aspirin treatment, potentially affecting the expression of genes in monocytes, twelve controls were subsequently treated with simvastatin and aspirin for 6 and 2 weeks, respectively. By whole genome expression arrays six genes were identified to have differential expression in the monocytes of patients versus controls; ABCA1, ABCG1 and RGS1 were downregulated in patients, whereas ADRB2, FOLR3 and GSTM1 were upregulated. Differential expression of all genes, apart from GSTM1, was confirmed by qPCR. Aspirin and statins altered gene expression of ABCG1 and ADBR2. All finding were validated in a second group of twenty four patients and controls. Differential expression of ABCA1, RSG1 and ADBR2 was replicated. In conclusion, we identified these 3 genes to be expressed differently in CAD cases which might play a role in the pathogenesis of atherosclerotic vascular disease.
Collapse
|
132
|
Jickling GC, Zhan X, Stamova B, Ander BP, Tian Y, Liu D, Sison SM, Verro P, Johnston SC, Sharp FR. Ischemic transient neurological events identified by immune response to cerebral ischemia. Stroke 2012; 43:1006-12. [PMID: 22308247 DOI: 10.1161/strokeaha.111.638577] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND PURPOSE Deciphering whether a transient neurological event (TNE) is of ischemic or nonischemic etiology can be challenging. Ischemia of cerebral tissue elicits an immune response in stroke and transient ischemic attack (TIA). This response, as detected by RNA expressed in immune cells, could potentially distinguish ischemic from nonischemic TNE. METHODS Analysis of 208 TIAs, ischemic strokes, controls, and TNE was performed. RNA from blood was processed on microarrays. TIAs (n=26) and ischemic strokes (n=94) were compared with controls (n=44) to identify differentially expressed genes (false discovery rate <0.05, fold change ≥1.2). Genes common to TIA and stroke were used predict ischemia in TIA diffusion-weighted imaging-positive/minor stroke (n=17), nonischemic TNE (n=13), and TNE of unclear etiology (n=14). RESULTS Seventy-four genes expressed in TIA were common to those in ischemic stroke. Functional pathways common to TIA and stroke related to activation of innate and adaptive immune systems, involving granulocytes and B cells. A prediction model using 26 of the 74 ischemia genes distinguished TIA and stroke subjects from control subjects with 89% sensitivity and specificity. In the validation cohort, 17 of 17 TIA diffusion-weighted imaging-positive/minor strokes were predicted to be ischemic, and 10 of 13 nonischemic TNE were predicted to be nonischemic. In TNE of unclear etiology, 71% were predicted to be ischemic. These subjects had higher ABCD(2) scores. CONCLUSIONS A common molecular response to ischemia in TIA and stroke was identified, relating to activation of innate and adaptive immune systems. TNE of ischemic etiology was identified based on gene profiles that may be of clinical use once validated.
Collapse
Affiliation(s)
- Glen C Jickling
- University of California at Davis, MIND Institute, 2805 50th Street, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Expression profiling of human immune cell subsets identifies miRNA-mRNA regulatory relationships correlated with cell type specific expression. PLoS One 2012; 7:e29979. [PMID: 22276136 PMCID: PMC3262799 DOI: 10.1371/journal.pone.0029979] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 12/07/2011] [Indexed: 01/01/2023] Open
Abstract
Blood consists of different cell populations with distinct functions and correspondingly, distinct gene expression profiles. In this study, global miRNA expression profiling was performed across a panel of nine human immune cell subsets (neutrophils, eosinophils, monocytes, B cells, NK cells, CD4 T cells, CD8 T cells, mDCs and pDCs) to identify cell-type specific miRNAs. mRNA expression profiling was performed on the same samples to determine if miRNAs specific to certain cell types down-regulated expression levels of their target genes. Six cell-type specific miRNAs (miR-143; neutrophil specific, miR-125; T cells and neutrophil specific, miR-500; monocyte and pDC specific, miR-150; lymphoid cell specific, miR-652 and miR-223; both myeloid cell specific) were negatively correlated with expression of their predicted target genes. These results were further validated using an independent cohort where similar immune cell subsets were isolated and profiled for both miRNA and mRNA expression. miRNAs which negatively correlated with target gene expression in both cohorts were identified as candidates for miRNA/mRNA regulatory pairs and were used to construct a cell-type specific regulatory network. miRNA/mRNA pairs formed two distinct clusters in the network corresponding to myeloid (nine miRNAs) and lymphoid lineages (two miRNAs). Several myeloid specific miRNAs targeted common genes including ABL2, EIF4A2, EPC1 and INO80D; these common targets were enriched for genes involved in the regulation of gene expression (p<9.0E-7). Those miRNA might therefore have significant further effect on gene expression by repressing the expression of genes involved in transcriptional regulation. The miRNA and mRNA expression profiles reported in this study form a comprehensive transcriptome database of various human blood cells and serve as a valuable resource for elucidating the role of miRNA mediated regulation in the establishment of immune cell identity.
Collapse
|
134
|
Abstract
In healthy individuals, T cells react against incoming pathogens, but remain tolerant to self-antigens, thereby preventing autoimmune reactions. CD4 regulatory T cells are major contributors in induction and maintenance of peripheral tolerance, but a regulatory role has been also reported for several subsets of CD8 T cells. To determine the molecular basis of peripheral CD8 T-cell tolerance, we exploited a double transgenic mouse model in which CD8 T cells are neonatally tolerized following interaction with a parenchymal self-antigen. These tolerant CD8 T cells have regulatory capacity and can suppress T cells in an antigen-specific manner during adulthood. Dickkopf-3 (DKK3) was found to be expressed in the tolerant CD8 T cells and to be essential for the observed CD8 T-cell tolerance. In vitro, genetic deletion of DKK3 or blocking with antibodies restored CD8 T-cell proliferation and IL-2 production in response to the tolerizing self-antigen. Moreover, exogenous DKK3 reduced CD8 T-cell reactivity. In vivo, abrogation of DKK3 function reversed tolerance, leading to eradication of tumors expressing the target antigen and to rejection of autologous skin grafts. Thus, our findings define DKK3 as a immune modulator with a crucial role for CD8 T-cell tolerance.
Collapse
|
135
|
Porwit A. Role of flow cytometry in diagnostics of myelodysplastic syndromes--beyond the WHO 2008 classification. Semin Diagn Pathol 2012; 28:273-82. [PMID: 22195405 DOI: 10.1053/j.semdp.2011.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiparameter flow cytometry (FCM) is an excellent method to follow the expression patterns of differentiation antigens using monoclonal antibodies to surface and cytoplasmic proteins. Although several authors described various aberrant immunophenotypic features in the bone marrow of patients with myelodysplastic syndromes (MDS), the World Health Organization 2008 classification recommended that, only if 3 or more phenotypic abnormalities are found involving 1 or more of the myeloid lineages can the aberrant FCM findings be considered suggestive of MDS. In the absence of conclusive morphologic and/or cytogenetic features, FCM abnormalities alone were considered not sufficient to establish MDS diagnosis and further follow-up of the patients was recommended. Review of the literature gives accumulating evidence that FCM has become an important part of the integrated diagnostic work-up of patients with suspected MDS. Several studies have also reported FCM findings significant for prognosis and therapy choice in MDS patients. Technical progress in multicolor FCM and new analysis programs, together with ongoing efforts to standardize the methodology, will make it possible to apply FCM in individual risk assessment and choice of best therapy for MDS patients.
Collapse
Affiliation(s)
- Anna Porwit
- Department of Laboratory Hematology, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
136
|
Abstract
This chapter summarizes current ideas about the intracellular signaling that drives platelet responses to vascular injury. After a brief overview of platelet activation intended to place the signaling pathways into context, the first section considers the early events of platelet activation leading up to integrin activation and platelet aggregation. The focus is on the G protein-mediated events utilized by agonists such as thrombin and ADP, and the tyrosine kinase-based signaling triggered by collagen. The second section considers the events that occur after integrin engagement, some of which are dependent on close physical contact between platelets. A third section addresses the regulatory events that help to avoid unprovoked or excessive platelet activation, after which the final section briefly considers individual variations in platelet reactivity and the role of platelet signaling in the innate immune response and embryonic development.
Collapse
Affiliation(s)
- Timothy J Stalker
- Departments of Medicine and Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
137
|
Bohne F, Martínez-Llordella M, Lozano JJ, Miquel R, Benítez C, Londoño MC, Manzia TM, Angelico R, Swinkels DW, Tjalsma H, López M, Abraldes JG, Bonaccorsi-Riani E, Jaeckel E, Taubert R, Pirenne J, Rimola A, Tisone G, Sánchez-Fueyo A. Intra-graft expression of genes involved in iron homeostasis predicts the development of operational tolerance in human liver transplantation. J Clin Invest 2012; 122:368-382. [PMID: 22156196 PMCID: PMC3248302 DOI: 10.1172/jci59411] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 10/26/2011] [Indexed: 12/13/2022] Open
Abstract
Following organ transplantation, lifelong immunosuppressive therapy is required to prevent the host immune system from destroying the allograft. This can cause severe side effects and increased recipient morbidity and mortality. Complete cessation of immunosuppressive drugs has been successfully accomplished in selected transplant recipients, providing proof of principle that operational allograft tolerance is attainable in clinical transplantation. The intra-graft molecular pathways associated with successful drug withdrawal, however, are not well defined. In this study, we analyzed sequential blood and liver tissue samples collected from liver transplant recipients enrolled in a prospective multicenter immunosuppressive drug withdrawal clinical trial. Before initiation of drug withdrawal, operationally tolerant and non-tolerant recipients differed in the intra-graft expression of genes involved in the regulation of iron homeostasis. Furthermore, as compared with non-tolerant recipients, operationally tolerant patients exhibited higher serum levels of hepcidin and ferritin and increased hepatocyte iron deposition. Finally, liver tissue gene expression measurements accurately predicted the outcome of immunosuppressive withdrawal in an independent set of patients. These results point to a critical role for iron metabolism in the regulation of intra-graft alloimmune responses in humans and provide a set of biomarkers to conduct drug-weaning trials in liver transplantation.
Collapse
Affiliation(s)
- Felix Bohne
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Marc Martínez-Llordella
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Juan-José Lozano
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Rosa Miquel
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Carlos Benítez
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - María-Carlota Londoño
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Tommaso-María Manzia
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Roberta Angelico
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Dorine W. Swinkels
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Harold Tjalsma
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Marta López
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Juan G. Abraldes
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Eliano Bonaccorsi-Riani
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Elmar Jaeckel
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Richard Taubert
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Antoni Rimola
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Giuseppe Tisone
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| | - Alberto Sánchez-Fueyo
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain.
Bioinformatics Platform, CIBEREHD, Barcelona, Spain.
Liver Transplant Unit, Surgical Clinic, University of Rome “Tor Vergata,” Rome, Italy.
Department of Laboratory Medicine, Laboratory of Genetic, Endocrine and Metabolic Diseases (830 LGEM), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany.
University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
138
|
Abstract
Although platelets are the smallest cells in the blood, they are implied in various processes ranging from immunology and oncology to thrombosis and hemostasis. Many large-scale screening programs, genome-wide association, and "omics" studies have generated lists of genes and loci that are probably involved in the formation or physiology of platelets under normal and pathologic conditions. This creates an increasing demand for new and improved model systems that allow functional assessment of the corresponding gene products in vivo. Such animal models not only render invaluable insight in the platelet biology, but in addition, provide improved test systems for the validation of newly developed anti-thrombotics. This review summarizes the most important models to generate transgenic platelets and to study their influence on platelet physiology in vivo. Here we focus on the zebrafish morpholino oligonucleotide technology, the (platelet-specific) knockout mouse, and the transplantation of genetically modified human or murine platelet progenitor cells in myelo-conditioned mice. The various strengths and pitfalls of these animal models are illustrated by recent examples from the platelet field. Finally, we highlight the latest developments in genetic engineering techniques and their possible application in platelet research.
Collapse
|
139
|
Rotival M, Zeller T, Wild PS, Maouche S, Szymczak S, Schillert A, Castagné R, Deiseroth A, Proust C, Brocheton J, Godefroy T, Perret C, Germain M, Eleftheriadis M, Sinning CR, Schnabel RB, Lubos E, Lackner KJ, Rossmann H, Münzel T, Rendon A, Consortium C, Erdmann J, Deloukas P, Hengstenberg C, Diemert P, Montalescot G, Ouwehand WH, Samani NJ, Schunkert H, Tregouet DA, Ziegler A, Goodall AH, Cambien F, Tiret L, Blankenberg S. Integrating genome-wide genetic variations and monocyte expression data reveals trans-regulated gene modules in humans. PLoS Genet 2011; 7:e1002367. [PMID: 22144904 PMCID: PMC3228821 DOI: 10.1371/journal.pgen.1002367] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/16/2011] [Indexed: 01/11/2023] Open
Abstract
One major expectation from the transcriptome in humans is to characterize the biological basis of associations identified by genome-wide association studies. So far, few cis expression quantitative trait loci (eQTLs) have been reliably related to disease susceptibility. Trans-regulating mechanisms may play a more prominent role in disease susceptibility. We analyzed 12,808 genes detected in at least 5% of circulating monocyte samples from a population-based sample of 1,490 European unrelated subjects. We applied a method of extraction of expression patterns-independent component analysis-to identify sets of co-regulated genes. These patterns were then related to 675,350 SNPs to identify major trans-acting regulators. We detected three genomic regions significantly associated with co-regulated gene modules. Association of these loci with multiple expression traits was replicated in Cardiogenics, an independent study in which expression profiles of monocytes were available in 758 subjects. The locus 12q13 (lead SNP rs11171739), previously identified as a type 1 diabetes locus, was associated with a pattern including two cis eQTLs, RPS26 and SUOX, and 5 trans eQTLs, one of which (MADCAM1) is a potential candidate for mediating T1D susceptibility. The locus 12q24 (lead SNP rs653178), which has demonstrated extensive disease pleiotropy, including type 1 diabetes, hypertension, and celiac disease, was associated to a pattern strongly correlating to blood pressure level. The strongest trans eQTL in this pattern was CRIP1, a known marker of cellular proliferation in cancer. The locus 12q15 (lead SNP rs11177644) was associated with a pattern driven by two cis eQTLs, LYZ and YEATS4, and including 34 trans eQTLs, several of them tumor-related genes. This study shows that a method exploiting the structure of co-expressions among genes can help identify genomic regions involved in trans regulation of sets of genes and can provide clues for understanding the mechanisms linking genome-wide association loci to disease.
Collapse
Affiliation(s)
- Maxime Rotival
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Tanja Zeller
- II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Philipp S. Wild
- II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Seraya Maouche
- Medizinische Klinik II, Universität Lübeck, Lübeck, Germany
| | - Silke Szymczak
- Institut für Medizinische Biometrie und Statistik, Universität Lübeck, Lübeck, Germany
| | - Arne Schillert
- Institut für Medizinische Biometrie und Statistik, Universität Lübeck, Lübeck, Germany
| | - Raphaele Castagné
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Arne Deiseroth
- II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Carole Proust
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Jessy Brocheton
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Tiphaine Godefroy
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Claire Perret
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Marine Germain
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Medea Eleftheriadis
- II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Christoph R. Sinning
- II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Renate B. Schnabel
- II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Edith Lubos
- II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Karl J. Lackner
- Institut für Klinische Chemie und Laboratoriumsmedizin, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Heidi Rossmann
- Institut für Klinische Chemie und Laboratoriumsmedizin, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Thomas Münzel
- II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Augusto Rendon
- Department of Haematology, University of Cambridge and National Health Service Blood and Transplant, Cambridge, United Kingdom
- MRC Biostatistics Unit, Cambridge, United Kingdom
| | | | | | - Panos Deloukas
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Christian Hengstenberg
- Klinik und Poliklinik für Innere Medizin II, Universität Regensburg, Regensburg, Germany
| | | | - Gilles Montalescot
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge and National Health Service Blood and Transplant, Cambridge, United Kingdom
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom
| | | | - David-Alexandre Tregouet
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Andreas Ziegler
- Institut für Medizinische Biometrie und Statistik, Universität Lübeck, Lübeck, Germany
| | - Alison H. Goodall
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom
| | - François Cambien
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Laurence Tiret
- INSERM UMRS 937, Pierre and Marie Curie University (UPMC, Paris 6) and Medical School, Paris, France
| | - Stefan Blankenberg
- II. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes-Gutenberg Universität Mainz, Mainz, Germany
| |
Collapse
|
140
|
Gieger C, Radhakrishnan A, Cvejic A, Tang W, Porcu E, Pistis G, Serbanovic-Canic J, Elling U, Goodall AH, Labrune Y, Lopez LM, Mägi R, Meacham S, Okada Y, Pirastu N, Sorice R, Teumer A, Voss K, Zhang W, Ramirez-Solis R, Bis JC, Ellinghaus D, Gögele M, Hottenga JJ, Langenberg C, Kovacs P, O'Reilly PF, Shin SY, Esko T, Hartiala J, Kanoni S, Murgia F, Parsa A, Stephens J, van der Harst P, Ellen van der Schoot C, Allayee H, Attwood A, Balkau B, Bastardot F, Basu S, Baumeister SE, Biino G, Bomba L, Bonnefond A, Cambien F, Chambers JC, Cucca F, D'Adamo P, Davies G, de Boer RA, de Geus EJC, Döring A, Elliott P, Erdmann J, Evans DM, Falchi M, Feng W, Folsom AR, Frazer IH, Gibson QD, Glazer NL, Hammond C, Hartikainen AL, Heckbert SR, Hengstenberg C, Hersch M, Illig T, Loos RJF, Jolley J, Khaw KT, Kühnel B, Kyrtsonis MC, Lagou V, Lloyd-Jones H, Lumley T, Mangino M, Maschio A, Mateo Leach I, McKnight B, Memari Y, Mitchell BD, Montgomery GW, Nakamura Y, Nauck M, Navis G, Nöthlings U, Nolte IM, Porteous DJ, Pouta A, Pramstaller PP, Pullat J, Ring SM, Rotter JI, Ruggiero D, Ruokonen A, Sala C, Samani NJ, Sambrook J, Schlessinger D, et alGieger C, Radhakrishnan A, Cvejic A, Tang W, Porcu E, Pistis G, Serbanovic-Canic J, Elling U, Goodall AH, Labrune Y, Lopez LM, Mägi R, Meacham S, Okada Y, Pirastu N, Sorice R, Teumer A, Voss K, Zhang W, Ramirez-Solis R, Bis JC, Ellinghaus D, Gögele M, Hottenga JJ, Langenberg C, Kovacs P, O'Reilly PF, Shin SY, Esko T, Hartiala J, Kanoni S, Murgia F, Parsa A, Stephens J, van der Harst P, Ellen van der Schoot C, Allayee H, Attwood A, Balkau B, Bastardot F, Basu S, Baumeister SE, Biino G, Bomba L, Bonnefond A, Cambien F, Chambers JC, Cucca F, D'Adamo P, Davies G, de Boer RA, de Geus EJC, Döring A, Elliott P, Erdmann J, Evans DM, Falchi M, Feng W, Folsom AR, Frazer IH, Gibson QD, Glazer NL, Hammond C, Hartikainen AL, Heckbert SR, Hengstenberg C, Hersch M, Illig T, Loos RJF, Jolley J, Khaw KT, Kühnel B, Kyrtsonis MC, Lagou V, Lloyd-Jones H, Lumley T, Mangino M, Maschio A, Mateo Leach I, McKnight B, Memari Y, Mitchell BD, Montgomery GW, Nakamura Y, Nauck M, Navis G, Nöthlings U, Nolte IM, Porteous DJ, Pouta A, Pramstaller PP, Pullat J, Ring SM, Rotter JI, Ruggiero D, Ruokonen A, Sala C, Samani NJ, Sambrook J, Schlessinger D, Schreiber S, Schunkert H, Scott J, Smith NL, Snieder H, Starr JM, Stumvoll M, Takahashi A, Tang WHW, Taylor K, Tenesa A, Lay Thein S, Tönjes A, Uda M, Ulivi S, van Veldhuisen DJ, Visscher PM, Völker U, Wichmann HE, Wiggins KL, Willemsen G, Yang TP, Hua Zhao J, Zitting P, Bradley JR, Dedoussis GV, Gasparini P, Hazen SL, Metspalu A, Pirastu M, Shuldiner AR, Joost van Pelt L, Zwaginga JJ, Boomsma DI, Deary IJ, Franke A, Froguel P, Ganesh SK, Jarvelin MR, Martin NG, Meisinger C, Psaty BM, Spector TD, Wareham NJ, Akkerman JWN, Ciullo M, Deloukas P, Greinacher A, Jupe S, Kamatani N, Khadake J, Kooner JS, Penninger J, Prokopenko I, Stemple D, Toniolo D, Wernisch L, Sanna S, Hicks AA, Rendon A, Ferreira MA, Ouwehand WH, Soranzo N. New gene functions in megakaryopoiesis and platelet formation. Nature 2011; 480:201-8. [PMID: 22139419 PMCID: PMC3335296 DOI: 10.1038/nature10659] [Show More Authors] [Citation(s) in RCA: 324] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 10/21/2011] [Indexed: 12/23/2022]
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.
Collapse
Affiliation(s)
- Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr 1, 85764 Neuherberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Serbanovic-Canic J, Cvejic A, Soranzo N, Stemple DL, Ouwehand WH, Freson K. Silencing of RhoA nucleotide exchange factor, ARHGEF3, reveals its unexpected role in iron uptake. Blood 2011; 118:4967-76. [PMID: 21715309 PMCID: PMC3208301 DOI: 10.1182/blood-2011-02-337295] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 06/13/2011] [Indexed: 11/20/2022] Open
Abstract
Genomewide association meta-analysis studies have identified > 100 independent genetic loci associated with blood cell indices, including volume and count of platelets and erythrocytes. Although several of these loci encode known regulators of hematopoiesis, the mechanism by which most sequence variants exert their effect on blood cell formation remains elusive. An example is the Rho guanine nucleotide exchange factor, ARHGEF3, which was previously implicated by genomewide association meta-analysis studies in bone cell biology. Here, we report on the unexpected role of ARHGEF3 in regulation of iron uptake and erythroid cell maturation. Although early erythroid differentiation progressed normally, silencing of arhgef3 in Danio rerio resulted in microcytic and hypochromic anemia. This was rescued by intracellular supplementation of iron, showing that arhgef3-depleted erythroid cells are fully capable of hemoglobinization. Disruption of the arhgef3 target, RhoA, also produced severe anemia, which was, again, corrected by iron injection. Moreover, silencing of ARHGEF3 in erythromyeloblastoid cells K562 showed that the uptake of transferrin was severely impaired. Taken together, this is the first study to provide evidence for ARHGEF3 being a regulator of transferrin uptake in erythroid cells, through activation of RHOA.
Collapse
Affiliation(s)
- Jovana Serbanovic-Canic
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
142
|
Castagné R, Zeller T, Rotival M, Szymczak S, Truong V, Schillert A, Trégouët DA, Münzel T, Ziegler A, Cambien F, Blankenberg S, Tiret L. Influence of sex and genetic variability on expression of X-linked genes in human monocytes. Genomics 2011; 98:320-6. [DOI: 10.1016/j.ygeno.2011.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 06/27/2011] [Accepted: 06/28/2011] [Indexed: 11/28/2022]
|
143
|
Woo AJ, Kim J, Xu J, Huang H, Cantor AB. Role of ZBP-89 in human globin gene regulation and erythroid differentiation. Blood 2011; 118:3684-93. [PMID: 21828133 PMCID: PMC3186340 DOI: 10.1182/blood-2011-03-341446] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 07/25/2011] [Indexed: 12/16/2022] Open
Abstract
The molecular mechanisms underlying erythroid-specific gene regulation remain incompletely understood. Closely spaced binding sites for GATA, NF-E2/maf, and CACCC interacting transcription factors play functionally important roles in globin and other erythroid-specific gene expression. We and others recently identified the CACCC-binding transcription factor ZBP-89 as a novel GATA-1 and NF-E2/mafK interacting partner. Here, we examined the role of ZBP-89 in human globin gene regulation and erythroid maturation using a primary CD34(+) cell ex vivo differentiation system. We show that ZBP-89 protein levels rise dramatically during human erythroid differentiation and that ZBP-89 occupies key cis-regulatory elements within the globin and other erythroid gene loci. ZBP-89 binding correlates strongly with RNA Pol II occupancy, active histone marks, and high-level gene expression. ZBP-89 physically associates with the histone acetyltransferases p300 and Gcn5/Trrap, and occupies common sites with Gcn5 within the human globin loci. Lentiviral short hairpin RNAs knockdown of ZBP-89 results in reduced Gcn5 occupancy, decreased acetylated histone 3 levels, lower globin and erythroid-specific gene expression, and impaired erythroid maturation. Addition of the histone deacetylase inhibitor valproic acid partially reverses the reduced globin gene expression. These findings reveal an activating role for ZBP-89 in human globin gene regulation and erythroid differentiation.
Collapse
Affiliation(s)
- Andrew J Woo
- Division of Pediatric Hematology/Oncology, Children's Hospital Boston and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
144
|
Lozano JJ, Pallier A, Martinez-Llordella M, Danger R, López M, Giral M, Londoño MC, Rimola A, Soulillou JP, Brouard S, Sánchez-Fueyo A. Comparison of transcriptional and blood cell-phenotypic markers between operationally tolerant liver and kidney recipients. Am J Transplant 2011; 11:1916-26. [PMID: 21827613 DOI: 10.1111/j.1600-6143.2011.03638.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A proportion of transplant recipients can spontaneously accept their grafts in the absence of immunosuppression (operational tolerance). Previous studies identified blood transcriptional and cell-phenotypic markers characteristic of either liver or kidney tolerant recipients. However, the small number of patients analyzed and the use of different transcriptional platforms hampered data interpretation. In this study we directly compared samples from kidney and liver tolerant recipients in order to identify potential similarities in immune-related parameters. Liver and kidney tolerant recipients differed in blood expression and B-cell immunophenotypic patterns and no significant overlaps were detectable between them. Whereas some recipients coincided in specific NK-related transcripts, this observation was not reproducible in all cohorts analyzed. Our results reveal that certain immune features, but not others, are consistently present across all cohorts of operationally tolerant recipients. This provides a set of reproducible biomarkers that should be explored in future large-scale immunomonitoring trials.
Collapse
Affiliation(s)
- J J Lozano
- Plataforma de Bioinformatica, CIBEREHD, Hospital Clinic, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Gremmels H, Fledderus JO, van Balkom BWM, Verhaar MC. Transcriptome analysis in endothelial progenitor cell biology. Antioxid Redox Signal 2011; 15:1029-42. [PMID: 20812873 DOI: 10.1089/ars.2010.3594] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The use of endothelial progenitor cells (EPCs) is a promising new treatment option for cardiovascular diseases. Many of the underlying mechanisms that result in an improvement of endothelial function in vivo remain poorly elucidated to this date, however. We summarize the current positions and potential applications of gene-expression profiling in the field of EPC biology. Based on our own and published gene-expression data, we demonstrate that gene-expression profiling can efficiently be used to characterize different EPC types. Furthermore, we highlight the potential of gene-expression profiling for the analysis of changes that EPCs undergo during culture and examine changes in gene transcription in diseased patients. Transcriptome profiling is a powerful tool for the characterization and functional analysis of EPCs in health and disease.
Collapse
Affiliation(s)
- Hendrik Gremmels
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
146
|
Liu P, Barb J, Woodhouse K, Taylor JG, Munson PJ, Raghavachari N. Transcriptome profiling and sequencing of differentiated human hematopoietic stem cells reveal lineage-specific expression and alternative splicing of genes. Physiol Genomics 2011; 43:1117-34. [PMID: 21828245 DOI: 10.1152/physiolgenomics.00099.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hematopoietic differentiation is strictly regulated by complex network of transcription factors that are controlled by ligands binding to cell surface receptors. Disruptions of the intricate sequences of transcriptional activation and suppression of multiple genes cause hematological diseases, such as leukemias, myelodysplastic syndromes, or myeloproliferative syndromes. From a clinical standpoint, deciphering the pattern of gene expression during hematopoiesis may help unravel disease-specific mechanisms in hematopoietic malignancies. Herein, we describe a human in vitro hematopoietic model system where lineage-specific differentiation of CD34(+) cells was accomplished using specific cytokines. Microarray and RNAseq-based whole transcriptome and exome analysis was performed on the differentiated erythropoietic, granulopoietic, and megakaryopoietic cells to delineate changes in expression of whole transcripts and exons. Analysis on the Human 1.0 ST exon arrays indicated differential expression of 172 genes (P < 0.0000001) and significant alternate splicing of 86 genes during differentiation. Pathway analysis identified these genes to be involved in Rac/RhoA signaling, Wnt/B-catenin signaling and alanine/aspartate metabolism. Comparison of the microarray data to next generation RNAseq analysis during erythroid differentiation demonstrated a high degree of correlation in gene (R = 0.72) and exon (R = 0.62) expression. Our data provide a molecular portrait of events that regulate differentiation of hematopoietic cells. Knowledge of molecular processes by which the cells acquire their cell-specific fate would be beneficial in developing cell-based therapies for human diseases.
Collapse
Affiliation(s)
- Poching Liu
- Genomics Core Facility, Genetics and Development Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
147
|
Vieira-de-Abreu A, Campbell RA, Weyrich AS, Zimmerman GA. Platelets: versatile effector cells in hemostasis, inflammation, and the immune continuum. Semin Immunopathol 2011; 34:5-30. [PMID: 21818701 DOI: 10.1007/s00281-011-0286-4] [Citation(s) in RCA: 230] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 07/20/2011] [Indexed: 12/28/2022]
Abstract
Platelets are chief effector cells in hemostasis. In addition, however, their specializations include activities and intercellular interactions that make them key effectors in inflammation and in the continuum of innate and adaptive immunity. This review focuses on the immune features of human platelets and platelets from experimental animals and on interactions between inflammatory, immune, and hemostatic activities of these anucleate but complex and versatile cells. The experimental findings and evidence for physiologic immune functions include previously unrecognized biologic characteristics of platelets and are paralleled by new evidence for unique roles of platelets in inflammatory, immune, and thrombotic diseases.
Collapse
Affiliation(s)
- Adriana Vieira-de-Abreu
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | | | | | |
Collapse
|
148
|
Hombrink P, Hadrup SR, Bakker A, Kester MGD, Falkenburg JHF, von dem Borne PA, Schumacher TNM, Heemskerk MHM. High-throughput identification of potential minor histocompatibility antigens by MHC tetramer-based screening: feasibility and limitations. PLoS One 2011; 6:e22523. [PMID: 21850230 PMCID: PMC3151248 DOI: 10.1371/journal.pone.0022523] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 06/22/2011] [Indexed: 11/28/2022] Open
Abstract
T-cell recognition of minor histocompatibility antigens (MiHA) plays an important role in the graft-versus-tumor (GVT) effect of allogeneic stem cell transplantation (allo-SCT). However, the number of MiHA identified to date remains limited, making clinical application of MiHA reactive T-cell infusion difficult. This study represents the first attempt of genome-wide prediction of MiHA, coupled to the isolation of T-cell populations that react with these antigens. In this unbiased high-throughput MiHA screen, both the possibilities and pitfalls of this approach were investigated. First, 973 polymorphic peptides expressed by hematopoietic stem cells were predicted and screened for HLA-A2 binding. Subsequently a set of 333 high affinity HLA-A2 ligands was identified and post transplantation samples from allo-SCT patients were screened for T-cell reactivity by a combination of pMHC-tetramer-based enrichment and multi-color flow cytometry. Using this approach, 71 peptide-reactive T-cell populations were generated. The isolation of a T-cell line specifically recognizing target cells expressing the MAP4K1IMA antigen demonstrates that identification of MiHA through this approach is in principle feasible. However, with the exception of the known MiHA HMHA1, none of the other T-cell populations that were generated demonstrated recognition of endogenously MiHA expressing target cells, even though recognition of peptide-loaded targets was often apparent. Collectively these results demonstrate the technical feasibility of high-throughput analysis of antigen-specific T-cell responses in small patient samples. However, the high-sensitivity of this approach requires the use of potential epitope sets that are not solely based on MHC binding, to prevent the frequent detection of T-cell responses that lack biological relevance.
Collapse
Affiliation(s)
- Pleun Hombrink
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Fehrmann RSN, Jansen RC, Veldink JH, Westra HJ, Arends D, Bonder MJ, Fu J, Deelen P, Groen HJM, Smolonska A, Weersma RK, Hofstra RMW, Buurman WA, Rensen S, Wolfs MGM, Platteel M, Zhernakova A, Elbers CC, Festen EM, Trynka G, Hofker MH, Saris CGJ, Ophoff RA, van den Berg LH, van Heel DA, Wijmenga C, te Meerman GJ, Franke L. Trans-eQTLs reveal that independent genetic variants associated with a complex phenotype converge on intermediate genes, with a major role for the HLA. PLoS Genet 2011; 7:e1002197. [PMID: 21829388 PMCID: PMC3150446 DOI: 10.1371/journal.pgen.1002197] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 06/06/2011] [Indexed: 12/19/2022] Open
Abstract
For many complex traits, genetic variants have been found associated. However, it is still mostly unclear through which downstream mechanism these variants cause these phenotypes. Knowledge of these intermediate steps is crucial to understand pathogenesis, while also providing leads for potential pharmacological intervention. Here we relied upon natural human genetic variation to identify effects of these variants on trans-gene expression (expression quantitative trait locus mapping, eQTL) in whole peripheral blood from 1,469 unrelated individuals. We looked at 1,167 published trait- or disease-associated SNPs and observed trans-eQTL effects on 113 different genes, of which we replicated 46 in monocytes of 1,490 different individuals and 18 in a smaller dataset that comprised subcutaneous adipose, visceral adipose, liver tissue, and muscle tissue. HLA single-nucleotide polymorphisms (SNPs) were 10-fold enriched for trans-eQTLs: 48% of the trans-acting SNPs map within the HLA, including ulcerative colitis susceptibility variants that affect plausible candidate genes AOAH and TRBV18 in trans. We identified 18 pairs of unlinked SNPs associated with the same phenotype and affecting expression of the same trans-gene (21 times more than expected, P<10−16). This was particularly pronounced for mean platelet volume (MPV): Two independent SNPs significantly affect the well-known blood coagulation genes GP9 and F13A1 but also C19orf33, SAMD14, VCL, and GNG11. Several of these SNPs have a substantially higher effect on the downstream trans-genes than on the eventual phenotypes, supporting the concept that the effects of these SNPs on expression seems to be much less multifactorial. Therefore, these trans-eQTLs could well represent some of the intermediate genes that connect genetic variants with their eventual complex phenotypic outcomes. Many genetic variants have been found associated with diseases. However, for many of these genetic variants, it remains unclear how they exert their effect on the eventual phenotype. We investigated genetic variants that are known to be associated with diseases and complex phenotypes and assessed whether these variants were also associated with gene expression levels in a set of 1,469 unrelated whole blood samples. For several diseases, such as type 1 diabetes and ulcerative colitis, we observed that genetic variants affect the expression of genes, not implicated before. For complex traits, such as mean platelet volume and mean corpuscular volume, we observed that independent genetic variants on different chromosomes influence the expression of exactly the same genes. For mean platelet volume, these genes include well-known blood coagulation genes but also genes with still unknown functions. These results indicate that, by systematically correlating genetic variation with gene expression levels, it is possible to identify downstream genes, which provide important avenues for further research.
Collapse
Affiliation(s)
- Rudolf S. N. Fehrmann
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Ritsert C. Jansen
- Groningen Bioinformatics Centre, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Haren, The Netherlands
| | - Jan H. Veldink
- Department of Neurology, Rudolf Magnus Institute of Neuroscience, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Harm-Jan Westra
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Danny Arends
- Groningen Bioinformatics Centre, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Haren, The Netherlands
| | - Marc Jan Bonder
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Patrick Deelen
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Harry J. M. Groen
- Department of Pulmonology, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Asia Smolonska
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Rinse K. Weersma
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen and University of Groningen, Groningen, The Netherlands
| | - Robert M. W. Hofstra
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Wim A. Buurman
- NUTRIM School for Nutrition, Toxicology, and Metabolism, Department of General Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sander Rensen
- NUTRIM School for Nutrition, Toxicology, and Metabolism, Department of General Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marcel G. M. Wolfs
- Department of Pathology and Medical Biology, Medical Biology Section, Molecular Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Mathieu Platteel
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Clara C. Elbers
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Eleanora M. Festen
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Gosia Trynka
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Marten H. Hofker
- Department of Pathology and Medical Biology, Medical Biology Section, Molecular Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Christiaan G. J. Saris
- Department of Neurology, Rudolf Magnus Institute of Neuroscience, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Roel A. Ophoff
- Department of Neurology, Rudolf Magnus Institute of Neuroscience, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Leonard H. van den Berg
- Department of Neurology, Rudolf Magnus Institute of Neuroscience, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - David A. van Heel
- Blizard Institute of Cell and Molecular Science, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Gerard J. te Meerman
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
- Blizard Institute of Cell and Molecular Science, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- * E-mail:
| |
Collapse
|
150
|
Thomas SG, Calaminus SDJ, Machesky LM, Alberts AS, Watson SP. G-protein coupled and ITAM receptor regulation of the formin FHOD1 through Rho kinase in platelets. J Thromb Haemost 2011; 9:1648-51. [PMID: 21605332 DOI: 10.1111/j.1538-7836.2011.04357.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|