Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Augusto Rendon is active.

Publication


Featured researches published by Augusto Rendon.


Nature Communications | 2015

Novel loci affecting iron homeostasis and their effects in individuals at risk for hemochromatosis (vol 5, 4926, 2014)

Beben Benyamin; Tonu Esko; Janina S. Ried; Aparna Radhakrishnan; Sita H. Vermeulen; Michela Traglia; Martin Goegele; Denise Anderson; Linda Broer; Clara Podmore; Jian'an Luan; Zoltán Kutalik; Serena Sanna; Peter van der Meer; Toshiko Tanaka; Fudi Wang; Harm-Jan Westra; Lude Franke; Evelin Mihailov; Lili Milani; Jonas Haelldin; Juliane Winkelmann; Thomas Meitinger; Joachim Thiery; Annette Peters; Melanie Waldenberger; Augusto Rendon; Jennifer Jolley; Jennifer Sambrook; Lambertus A. Kiemeney

Corrigendum: Novel loci affecting iron homeostasis and their effects in individuals at risk for hemochromatosis


Nature Genetics | 2009

A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium.

Nicole Soranzo; Tim D. Spector; Massimo Mangino; Brigitte Kühnel; Augusto Rendon; Alexander Teumer; Christina Willenborg; Benjamin J. Wright; Li Chen; Mingyao Li; Perttu Salo; Benjamin F. Voight; Philippa Burns; Roman A. Laskowski; Yali Xue; Stephan Menzel; David Altshuler; John R. Bradley; Suzannah Bumpstead; Mary-Susan Burnett; Joseph M. Devaney; Angela Döring; Roberto Elosua; Stephen E. Epstein; Wendy N. Erber; Mario Falchi; Stephen F. Garner; Mohammed J. R. Ghori; Alison H. Goodall; Rhian Gwilliam

The number and volume of cells in the blood affect a wide range of disorders including cancer and cardiovascular, metabolic, infectious and immune conditions. We consider here the genetic variation in eight clinically relevant hematological parameters, including hemoglobin levels, red and white blood cell counts and platelet counts and volume. We describe common variants within 22 genetic loci reproducibly associated with these hematological parameters in 13,943 samples from six European population-based studies, including 6 associated with red blood cell parameters, 15 associated with platelet parameters and 1 associated with total white blood cell count. We further identified a long-range haplotype at 12q24 associated with coronary artery disease and myocardial infarction in 9,479 cases and 10,527 controls. We show that this haplotype demonstrates extensive disease pleiotropy, as it contains known risk loci for type 1 diabetes, hypertension and celiac disease and has been spread by a selective sweep specific to European and geographically nearby populations.


The Lancet | 2012

Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies.

Nadeem Sarwar; Adam S. Butterworth; Daniel F. Freitag; John Gregson; Peter Willeit; Donal N. Gorman; Pei Gao; Danish Saleheen; Augusto Rendon; Christopher P. Nelson; Peter S. Braund; Alistair S. Hall; Daniel I. Chasman; Anne Tybjærg-Hansen; John Chambers; Emelia J. Benjamin; Paul W. Franks; Robert Clarke; Arthur A. M. Wilde; Mieke D. Trip; Maristella Steri; Jacqueline C. M. Witteman; Lu Qi; C. Ellen van der Schoot; Ulf de Faire; Jeanette Erdmann; H. M. Stringham; Wolfgang Koenig; Daniel J. Rader; David Melzer

Summary Background Persistent inflammation has been proposed to contribute to various stages in the pathogenesis of cardiovascular disease. Interleukin-6 receptor (IL6R) signalling propagates downstream inflammation cascades. To assess whether this pathway is causally relevant to coronary heart disease, we studied a functional genetic variant known to affect IL6R signalling. Methods In a collaborative meta-analysis, we studied Asp358Ala (rs2228145) in IL6R in relation to a panel of conventional risk factors and inflammation biomarkers in 125 222 participants. We also compared the frequency of Asp358Ala in 51 441 patients with coronary heart disease and in 136 226 controls. To gain insight into possible mechanisms, we assessed Asp358Ala in relation to localised gene expression and to postlipopolysaccharide stimulation of interleukin 6. Findings The minor allele frequency of Asp358Ala was 39%. Asp358Ala was not associated with lipid concentrations, blood pressure, adiposity, dysglycaemia, or smoking (p value for association per minor allele ≥0·04 for each). By contrast, for every copy of 358Ala inherited, mean concentration of IL6R increased by 34·3% (95% CI 30·4–38·2) and of interleukin 6 by 14·6% (10·7–18·4), and mean concentration of C-reactive protein was reduced by 7·5% (5·9–9·1) and of fibrinogen by 1·0% (0·7–1·3). For every copy of 358Ala inherited, risk of coronary heart disease was reduced by 3·4% (1·8–5·0). Asp358Ala was not related to IL6R mRNA levels or interleukin-6 production in monocytes. Interpretation Large-scale human genetic and biomarker data are consistent with a causal association between IL6R-related pathways and coronary heart disease. Funding British Heart Foundation; UK Medical Research Council; UK National Institute of Health Research, Cambridge Biomedical Research Centre; BUPA Foundation.


Nature Genetics | 2009

Multiple loci influence erythrocyte phenotypes in the CHARGE Consortium

Santhi K. Ganesh; Neil A. Zakai; Frank J. A. van Rooij; Nicole Soranzo; Albert V. Smith; Michael A. Nalls; Ming-Huei Chen; Anna Köttgen; Nicole L. Glazer; Abbas Dehghan; Brigitte Kühnel; Thor Aspelund; Qiong Yang; Toshiko Tanaka; Andrew E. Jaffe; Joshua C. Bis; Germaine C. Verwoert; Alexander Teumer; Caroline S. Fox; Jack M. Guralnik; Georg B. Ehret; Kenneth Rice; Janine F. Felix; Augusto Rendon; Gudny Eiriksdottir; Daniel Levy; Kushang V. Patel; Eric Boerwinkle; Jerome I. Rotter; Albert Hofman

Measurements of erythrocytes within the blood are important clinical traits and can indicate various hematological disorders. We report here genome-wide association studies (GWAS) for six erythrocyte traits, including hemoglobin concentration (Hb), hematocrit (Hct), mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC) and red blood cell count (RBC). We performed an initial GWAS in cohorts of the CHARGE Consortium totaling 24,167 individuals of European ancestry and replication in additional independent cohorts of the HaemGen Consortium totaling 9,456 individuals. We identified 23 loci significantly associated with these traits in a meta-analysis of the discovery and replication cohorts (combined P values ranging from 5 × 10−8 to 7 × 10−86). Our findings include loci previously associated with these traits (HBS1L-MYB, HFE, TMPRSS6, TFR2, SPTA1) as well as new associations (EPO, TFRC, SH2B3 and 15 other loci). This study has identified new determinants of erythrocyte traits, offering insight into common variants underlying variation in erythrocyte measures.


Nature Genetics | 2011

Exome sequencing identifies NBEAL2 as the causative gene for gray platelet syndrome

Cornelis A. Albers; Ana Cvejic; Rémi Favier; Evelien E Bouwmans; Marie-Christine Alessi; Paul Bertone; Gregory Jordan; Ross Kettleborough; Graham Kiddle; Myrto Kostadima; Randy J. Read; Botond Sipos; Suthesh Sivapalaratnam; Peter A. Smethurst; Jonathan Stephens; Katrin Voss; Alan T. Nurden; Augusto Rendon; Paquita Nurden; Willem H. Ouwehand

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


Science | 2014

Transcriptional diversity during lineage commitment of human blood progenitors

Lu Chen; Myrto Kostadima; Joost H.A. Martens; Giovanni Canu; Sara P. Garcia; Ernest Turro; Kate Downes; Iain C. Macaulay; Ewa Bielczyk-Maczyńska; Sophia Coe; Samantha Farrow; Pawan Poudel; Frances Burden; Sjoert B. G. Jansen; William Astle; Antony P. Attwood; Tadbir K. Bariana; Bernard de Bono; Alessandra Breschi; John Chambers; Fizzah Choudry; Laura Clarke; Paul Coupland; Martijn van der Ent; Wendy N. Erber; Joop H. Jansen; Rémi Favier; Matthew Fenech; Nicola S. Foad; Kathleen Freson

Introduction Blood production in humans culminates in the daily release of around 1011 cells into the circulation, mainly platelets and red blood cells. All blood cells originate from a minute population of hematopoietic stem cells (HSCs) that expands and differentiates into progenitor cells with increasingly restricted lineage choice. Characterizing alternative splicing events involved in hematopoiesis is critical for interpreting the effects of mutations leading to inherited disorders and blood cancers and for the rational design of strategies to advance transplantation and regenerative medicine. Overview of methodology. RNA-sequencing reads from human blood progenitors [opaque cells in (A)] were mapped to the transcriptome to quantify gene and transcript expression. Reads were also mapped to the genome to identify novel splice junctions and characterize alternative splicing events (B). Rationale To address this, we explored the transcriptional diversity of human blood progenitors by sequencing RNA from six progenitor and two precursor populations representing the classical myeloid commitment stages of hematopoiesis and the main lymphoid stage. Data were aligned to the human reference transcriptome and genome to quantify known transcript isoforms and to identify novel splicing events, respectively. We used Bayesian polytomous model selection to classify transcripts into distinct expression patterns across the three cell types that comprise each differentiation step. Results We identified extensive transcriptional changes involving 6711 genes and 10,724 transcripts and validated a number of these. Many of the changes at the transcript isoform level did not result in significant changes at the gene expression level. Moreover, we identified transcripts unique to each of the progenitor populations, observing enrichment in non–protein-coding elements at the early stages of differentiation. We discovered 7881 novel splice junctions and 2301 differentially used alternative splicing events, enriched in genes involved in regulatory processes and often resulting in the gain or loss of functional domains. Of the alternative splice sites displaying differential usage, 73% resulted in exon-skipping events involving at least one protein domain (38.5%) or introducing a premature stop codon (26%). Enrichment analysis of RNA-binding motifs provided insights into the regulation of cell type–specific splicing events. To demonstrate the importance of specific isoforms in driving lineage fating events, we investigated the role of a transcription factor highlighted by our analyses. Our data show that nuclear factor I/B (NFIB) is highly expressed in megakaryocytes and that it is transcribed from an unannotated transcription start site preceding a novel exon. The novel NFIB isoform lacks the DNA binding/dimerization domain and therefore is unable to interact with its binding partner, NFIC. We further show that NFIB and NFIC are important in megakaryocyte differentiation. Conclusion We produced a quantitative catalog of transcriptional changes and splicing events representing the early progenitors of human blood. Our analyses unveil a previously undetected layer of regulation affecting cell fating, which involves transcriptional isoforms switching without noticeable changes at the gene level and resulting in the gain or loss of protein functions. A BLUEPRINT of immune cell development To determine the epigenetic mechanisms that direct blood cells to develop into the many components of our immune system, the BLUEPRINT consortium examined the regulation of DNA and RNA transcription to dissect the molecular traits that govern blood cell differentiation. By inducing immune responses, Saeed et al. document the epigenetic changes in the genome that underlie immune cell differentiation. Cheng et al. demonstrate that trained monocytes are highly dependent on the breakdown of sugars in the presence of oxygen, which allows cells to produce the energy needed to mount an immune response. Chen et al. examine RNA transcripts and find that specific cell lineages use RNA transcripts of different length and composition (isoforms) to form proteins. Together, the studies reveal how epigenetic effects can drive the development of blood cells involved in the immune system. Science, this issue 10.1126/science.1251086, 10.1126/science.1250684, 10.1126/science.1251033 RNA sequencing identifies how different cell fate decisions are made during blood cell differentiation. Blood cells derive from hematopoietic stem cells through stepwise fating events. To characterize gene expression programs driving lineage choice, we sequenced RNA from eight primary human hematopoietic progenitor populations representing the major myeloid commitment stages and the main lymphoid stage. We identified extensive cell type–specific expression changes: 6711 genes and 10,724 transcripts, enriched in non–protein-coding elements at early stages of differentiation. In addition, we found 7881 novel splice junctions and 2301 differentially used alternative splicing events, enriched in genes involved in regulatory processes. We demonstrated experimentally cell-specific isoform usage, identifying nuclear factor I/B (NFIB) as a regulator of megakaryocyte maturation—the platelet precursor. Our data highlight the complexity of fating events in closely related progenitor populations, the understanding of which is essential for the advancement of transplantation and regenerative medicine.


Blood | 2009

A novel variant on chromosome 7q22.3 associated with mean platelet volume, counts, and function.

Nicole Soranzo; Augusto Rendon; Christian Gieger; Chris I. Jones; Nicholas A. Watkins; Stephan Menzel; Angela Döring; Jonathan Stephens; Holger Prokisch; Wendy N. Erber; Simon Potter; Sarah L. Bray; Philippa Burns; Jennifer Jolley; Mario Falchi; Brigitte Kühnel; Jeanette Erdmann; Heribert Schunkert; Nilesh J. Samani; Thomas Illig; Stephen F. Garner; Angela Rankin; Christa Meisinger; John R. Bradley; Swee Lay Thein; Alison H. Goodall; Tim D. Spector; Panos Deloukas; Willem H. Ouwehand

Mean platelet volume (MPV) and platelet count (PLT) are highly heritable and tightly regulated traits. We performed a genome-wide association study for MPV and identified one SNP, rs342293, as having highly significant and reproducible association with MPV (per-G allele effect 0.016 +/- 0.001 log fL; P < 1.08 x 10(-24)) and PLT (per-G effect -4.55 +/- 0.80 10(9)/L; P < 7.19 x 10(-8)) in 8586 healthy subjects. Whole-genome expression analysis in the 1-MB region showed a significant association with platelet transcript levels for PIK3CG (n = 35; P = .047). The G allele at rs342293 was also associated with decreased binding of annexin V to platelets activated with collagen-related peptide (n = 84; P = .003). The region 7q22.3 identifies the first QTL influencing platelet volume, counts, and function in healthy subjects. Notably, the association signal maps to a chromosome region implicated in myeloid malignancies, indicating this site as an important regulatory site for hematopoiesis. The identification of loci regulating MPV by this and other studies will increase our insight in the processes of megakaryopoiesis and proplatelet formation, and it may aid the identification of genes that are somatically mutated in essential thrombocytosis.


PLOS Genetics | 2011

Multiple Loci Are Associated with White Blood Cell Phenotypes

Michael A. Nalls; David Couper; Toshiko Tanaka; Frank J. A. van Rooij; Ming-Huei Chen; Albert V. Smith; Daniela Toniolo; Neil A. Zakai; Qiong Yang; Andreas Greinacher; Andrew R. Wood; Melissa Garcia; Paolo Gasparini; Yongmei Liu; Thomas Lumley; Aaron R. Folsom; Alex P. Reiner; Christian Gieger; Vasiliki Lagou; Janine F. Felix; Henry Völzke; Natalia Gouskova; Alessandro Biffi; Angela Döring; Uwe Völker; Sean Chong; Kerri L. Wiggins; Augusto Rendon; Abbas Dehghan; Matt Moore

White blood cell (WBC) count is a common clinical measure from complete blood count assays, and it varies widely among healthy individuals. Total WBC count and its constituent subtypes have been shown to be moderately heritable, with the heritability estimates varying across cell types. We studied 19,509 subjects from seven cohorts in a discovery analysis, and 11,823 subjects from ten cohorts for replication analyses, to determine genetic factors influencing variability within the normal hematological range for total WBC count and five WBC subtype measures. Cohort specific data was supplied by the CHARGE, HeamGen, and INGI consortia, as well as independent collaborative studies. We identified and replicated ten associations with total WBC count and five WBC subtypes at seven different genomic loci (total WBC count—6p21 in the HLA region, 17q21 near ORMDL3, and CSF3; neutrophil count—17q21; basophil count- 3p21 near RPN1 and C3orf27; lymphocyte count—6p21, 19p13 at EPS15L1; monocyte count—2q31 at ITGA4, 3q21, 8q24 an intergenic region, 9q31 near EDG2), including three previously reported associations and seven novel associations. To investigate functional relationships among variants contributing to variability in the six WBC traits, we utilized gene expression- and pathways-based analyses. We implemented gene-clustering algorithms to evaluate functional connectivity among implicated loci and showed functional relationships across cell types. Gene expression data from whole blood was utilized to show that significant biological consequences can be extracted from our genome-wide analyses, with effect estimates for significant loci from the meta-analyses being highly corellated with the proximal gene expression. In addition, collaborative efforts between the groups contributing to this study and related studies conducted by the COGENT and RIKEN groups allowed for the examination of effect homogeneity for genome-wide significant associations across populations of diverse ancestral backgrounds.


PLOS Genetics | 2011

Integrating genome-wide genetic variations and monocyte expression data reveals trans-regulated gene modules in humans.

Maxime Rotival; Tanja Zeller; Philipp S. Wild; Seraya Maouche; Silke Szymczak; Arne Schillert; Raphaële Castagné; Arne Deiseroth; Carole Proust; Jessy Brocheton; Tiphaine Godefroy; Claire Perret; Marine Germain; Medea Eleftheriadis; Christoph Sinning; Renate B. Schnabel; Edith Lubos; Karl J. Lackner; Heidi Rossmann; Thomas Münzel; Augusto Rendon; Jeanette Erdmann; Panos Deloukas; Christian Hengstenberg; Patrick Diemert; Gilles Montalescot; Willem H. Ouwehand; Nilesh J. Samani; Heribert Schunkert; David-Alexandre Trégouët

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.


Cell | 2016

Genetic Drivers of Epigenetic and Transcriptional Variation in Human Immune Cells

Lu Chen; Bing Ge; Francesco Paolo Casale; Louella Vasquez; Tony Kwan; Diego Garrido-Martín; Stephen Watt; Ying Yan; Kousik Kundu; Simone Ecker; Avik Datta; David C. Richardson; Frances Burden; Daniel Mead; Alice L. Mann; José María Fernández; Sophia Rowlston; Steven P. Wilder; Samantha Farrow; Xiaojian Shao; John J. Lambourne; Adriana Redensek; Cornelis A. Albers; Vyacheslav Amstislavskiy; Sofie Ashford; Kim Berentsen; Lorenzo Bomba; Guillaume Bourque; David Bujold; Stephan Busche

Summary Characterizing the multifaceted contribution of genetic and epigenetic factors to disease phenotypes is a major challenge in human genetics and medicine. We carried out high-resolution genetic, epigenetic, and transcriptomic profiling in three major human immune cell types (CD14+ monocytes, CD16+ neutrophils, and naive CD4+ T cells) from up to 197 individuals. We assess, quantitatively, the relative contribution of cis-genetic and epigenetic factors to transcription and evaluate their impact as potential sources of confounding in epigenome-wide association studies. Further, we characterize highly coordinated genetic effects on gene expression, methylation, and histone variation through quantitative trait locus (QTL) mapping and allele-specific (AS) analyses. Finally, we demonstrate colocalization of molecular trait QTLs at 345 unique immune disease loci. This expansive, high-resolution atlas of multi-omics changes yields insights into cell-type-specific correlation between diverse genomic inputs, more generalizable correlations between these inputs, and defines molecular events that may underpin complex disease risk.

Collaboration


Dive into the Augusto Rendon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicole Soranzo

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Panos Deloukas

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge