Network


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

Hotspot


Dive into the research topics where Manuel A. Rivas is active.

Publication


Featured researches published by Manuel A. Rivas.


Nature Genetics | 2011

A framework for variation discovery and genotyping using next-generation DNA sequencing data

Mark A. DePristo; Eric Banks; Ryan Poplin; Kiran Garimella; Jared Maguire; Christopher Hartl; Anthony A. Philippakis; Guillermo Del Angel; Manuel A. Rivas; Matt Hanna; Aaron McKenna; Timothy Fennell; Andrew Kernytsky; Andrey Sivachenko; Kristian Cibulskis; Stacey B. Gabriel; David Altshuler; Mark J. Daly

Recent advances in sequencing technology make it possible to comprehensively catalog genetic variation in population samples, creating a foundation for understanding human disease, ancestry and evolution. The amounts of raw data produced are prodigious, and many computational steps are required to translate this output into high-quality variant calls. We present a unified analytic framework to discover and genotype variation among multiple samples simultaneously that achieves sensitive and specific results across five sequencing technologies and three distinct, canonical experimental designs. Our process includes (i) initial read mapping; (ii) local realignment around indels; (iii) base quality score recalibration; (iv) SNP discovery and genotyping to find all potential variants; and (v) machine learning to separate true segregating variation from machine artifacts common to next-generation sequencing technologies. We here discuss the application of these tools, instantiated in the Genome Analysis Toolkit, to deep whole-genome, whole-exome capture and multi-sample low-pass (∼4×) 1000 Genomes Project datasets.


Nature | 2016

Analysis of protein-coding genetic variation in 60,706 humans

Monkol Lek; Konrad J. Karczewski; Eric Vallabh Minikel; Kaitlin E. Samocha; Eric Banks; Timothy Fennell; Anne H. O’Donnell-Luria; James S. Ware; Andrew Hill; Beryl B. Cummings; Taru Tukiainen; Daniel P. Birnbaum; Jack A. Kosmicki; Laramie Duncan; Karol Estrada; Fengmei Zhao; James Zou; Emma Pierce-Hoffman; Joanne Berghout; David Neil Cooper; Nicole Deflaux; Mark A. DePristo; Ron Do; Jason Flannick; Menachem Fromer; Laura Gauthier; Jackie Goldstein; Namrata Gupta; Daniel P. Howrigan; Adam Kiezun

Large-scale reference data sets of human genetic variation are critical for the medical and functional interpretation of DNA sequence changes. Here we describe the aggregation and analysis of high-quality exome (protein-coding region) DNA sequence data for 60,706 individuals of diverse ancestries generated as part of the Exome Aggregation Consortium (ExAC). This catalogue of human genetic diversity contains an average of one variant every eight bases of the exome, and provides direct evidence for the presence of widespread mutational recurrence. We have used this catalogue to calculate objective metrics of pathogenicity for sequence variants, and to identify genes subject to strong selection against various classes of mutation; identifying 3,230 genes with near-complete depletion of predicted protein-truncating variants, with 72% of these genes having no currently established human disease phenotype. Finally, we demonstrate that these data can be used for the efficient filtering of candidate disease-causing variants, and for the discovery of human ‘knockout’ variants in protein-coding genes.


Nature | 2013

Transcriptome and genome sequencing uncovers functional variation in humans.

Tuuli Lappalainen; Michael Sammeth; Marc R. Friedländer; Peter A. C. 't Hoen; Jean Monlong; Manuel A. Rivas; Mar Gonzàlez-Porta; Natalja Kurbatova; Thasso Griebel; Pedro G. Ferreira; Matthias Barann; Thomas Wieland; Liliana Greger; M. van Iterson; Jonas Carlsson Almlöf; Paolo Ribeca; Irina Pulyakhina; Daniela Esser; Thomas Giger; Andrew Tikhonov; Marc Sultan; G. Bertier; Daniel G. MacArthur; Monkol Lek; Esther Lizano; Henk P. J. Buermans; Ismael Padioleau; Thomas Schwarzmayr; Olof Karlberg; Halit Ongen

Genome sequencing projects are discovering millions of genetic variants in humans, and interpretation of their functional effects is essential for understanding the genetic basis of variation in human traits. Here we report sequencing and deep analysis of messenger RNA and microRNA from lymphoblastoid cell lines of 462 individuals from the 1000 Genomes Project—the first uniformly processed high-throughput RNA-sequencing data from multiple human populations with high-quality genome sequences. We discover extremely widespread genetic variation affecting the regulation of most genes, with transcript structure and expression level variation being equally common but genetically largely independent. Our characterization of causal regulatory variation sheds light on the cellular mechanisms of regulatory and loss-of-function variation, and allows us to infer putative causal variants for dozens of disease-associated loci. Altogether, this study provides a deep understanding of the cellular mechanisms of transcriptome variation and of the landscape of functional variants in the human genome.


Nature Genetics | 2011

Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease

Manuel A. Rivas; Mélissa Beaudoin; Agnès Gardet; Christine Stevens; Yashoda Sharma; Clarence K. Zhang; Gabrielle Boucher; Stephan Ripke; David Ellinghaus; Noël P. Burtt; Timothy Fennell; Andrew Kirby; Anna Latiano; Philippe Goyette; Todd Green; Jonas Halfvarson; Talin Haritunians; Joshua M. Korn; Finny Kuruvilla; Caroline Lagacé; Benjamin M. Neale; Ken Sin Lo; Phil Schumm; Leif Törkvist; Marla Dubinsky; Steven R. Brant; Mark S. Silverberg; Richard H. Duerr; David Altshuler; Stacey Gabriel

More than 1,000 susceptibility loci have been identified through genome-wide association studies (GWAS) of common variants; however, the specific genes and full allelic spectrum of causal variants underlying these findings have not yet been defined. Here we used pooled next-generation sequencing to study 56 genes from regions associated with Crohns disease in 350 cases and 350 controls. Through follow-up genotyping of 70 rare and low-frequency protein-altering variants in nine independent case-control series (16,054 Crohns disease cases, 12,153 ulcerative colitis cases and 17,575 healthy controls), we identified four additional independent risk factors in NOD2, two additional protective variants in IL23R, a highly significant association with a protective splice variant in CARD9 (P < 1 × 10−16, odds ratio ≈ 0.29) and additional associations with coding variants in IL18RAP, CUL2, C1orf106, PTPN22 and MUC19. We extend the results of successful GWAS by identifying new, rare and probably functional variants that could aid functional experiments and predictive models.


PLOS Genetics | 2011

Testing for an Unusual Distribution of Rare Variants

Benjamin M. Neale; Manuel A. Rivas; Benjamin F. Voight; David Altshuler; Bernie Devlin; Marju Orho-Melander; Sekar Kathiresan; Shaun Purcell; Kathryn Roeder; Mark J. Daly

Technological advances make it possible to use high-throughput sequencing as a primary discovery tool of medical genetics, specifically for assaying rare variation. Still this approach faces the analytic challenge that the influence of very rare variants can only be evaluated effectively as a group. A further complication is that any given rare variant could have no effect, could increase risk, or could be protective. We propose here the C-alpha test statistic as a novel approach for testing for the presence of this mixture of effects across a set of rare variants. Unlike existing burden tests, C-alpha, by testing the variance rather than the mean, maintains consistent power when the target set contains both risk and protective variants. Through simulations and analysis of case/control data, we demonstrate good power relative to existing methods that assess the burden of rare variants in individuals.


Nature | 2015

Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction

Ron Do; Nathan O. Stitziel; Hong-Hee Won; Anders Jørgensen; Stefano Duga; Pier Angelica Merlini; Adam Kiezun; Martin Farrall; Anuj Goel; Or Zuk; Illaria Guella; Rosanna Asselta; Leslie A. Lange; Gina M. Peloso; Paul L. Auer; Domenico Girelli; Nicola Martinelli; Deborah N. Farlow; Mark A. DePristo; Robert Roberts; Alex Stewart; Danish Saleheen; John Danesh; Stephen E. Epstein; Suthesh Sivapalaratnam; G. Kees Hovingh; John J. P. Kastelein; Nilesh J. Samani; Heribert Schunkert; Jeanette Erdmann

Summary Myocardial infarction (MI), a leading cause of death around the world, displays a complex pattern of inheritance1,2. When MI occurs early in life, the role of inheritance is substantially greater1. Previously, rare mutations in low-density lipoprotein (LDL) genes have been shown to contribute to MI risk in individual families3–8 whereas common variants at more than 45 loci have been associated with MI risk in the population9–15. Here, we evaluate the contribution of rare mutations to MI risk in the population. We sequenced the protein-coding regions of 9,793 genomes from patients with MI at an early age (≤50 years in males and ≤60 years in females) along with MI-free controls. We identified two genes where rare coding-sequence mutations were more frequent in cases versus controls at exome-wide significance. At low-density lipoprotein receptor (LDLR), carriers of rare, damaging mutations (3.1% of cases versus 1.3% of controls) were at 2.4-fold increased risk for MI; carriers of null alleles at LDLR were at even higher risk (13-fold difference). This sequence-based estimate of the proportion of early MI cases due to LDLR mutations is remarkably similar to an estimate made more than 40 years ago using total cholesterol16. At apolipoprotein A-V (APOA5), carriers of rare nonsynonymous mutations (1.4% of cases versus 0.6% of controls) were at 2.2-fold increased risk for MI. When compared with non-carriers, LDLR mutation carriers had higher plasma LDL cholesterol whereas APOA5 mutation carriers had higher plasma triglycerides. Recent evidence has connected MI risk with coding sequence mutations at two genes functionally related to APOA5, namely lipoprotein lipase15,17 and apolipoprotein C318,19. When combined, these observations suggest that, beyond LDL cholesterol, disordered metabolism of triglyceride-rich lipoproteins contributes to MI risk.


Nature Genetics | 2010

High-throughput, pooled sequencing identifies mutations in NUBPL and FOXRED1 in human complex I deficiency

Sarah E. Calvo; Elena J. Tucker; Alison G. Compton; Denise M. Kirby; Gabriel Crawford; Noël P. Burtt; Manuel A. Rivas; Candace Guiducci; Damien L. Bruno; Olga Goldberger; Michelle C Redman; Esko Wiltshire; Callum Wilson; David Altshuler; Stacey Gabriel; Mark J. Daly; David R. Thorburn; Vamsi K. Mootha

Discovering the molecular basis of mitochondrial respiratory chain disease is challenging given the large number of both mitochondrial and nuclear genes that are involved. We report a strategy of focused candidate gene prediction, high-throughput sequencing and experimental validation to uncover the molecular basis of mitochondrial complex I disorders. We created seven pools of DNA from a cohort of 103 cases and 42 healthy controls and then performed deep sequencing of 103 candidate genes to identify 151 rare variants that were predicted to affect protein function. We established genetic diagnoses in 13 of 60 previously unsolved cases using confirmatory experiments, including cDNA complementation to show that mutations in NUBPL and FOXRED1 can cause complex I deficiency. Our study illustrates how large-scale sequencing, coupled with functional prediction and experimental validation, can be used to identify causal mutations in individual cases.


PLOS Genetics | 2008

Genetic Analysis of Human Traits In Vitro: Drug Response and Gene Expression in Lymphoblastoid Cell Lines

Edwin Choy; Roman Yelensky; Sasha Bonakdar; Robert M. Plenge; Richa Saxena; Philip L. De Jager; Stanley Y. Shaw; Cara S Wolfish; Jacqueline M. Slavik; Chris Cotsapas; Manuel A. Rivas; Emmanouil T. Dermitzakis; Ellen Cahir-McFarland; Elliott Kieff; David A. Hafler; Mark J. Daly; David Altshuler

Lymphoblastoid cell lines (LCLs), originally collected as renewable sources of DNA, are now being used as a model system to study genotype–phenotype relationships in human cells, including searches for QTLs influencing levels of individual mRNAs and responses to drugs and radiation. In the course of attempting to map genes for drug response using 269 LCLs from the International HapMap Project, we evaluated the extent to which biological noise and non-genetic confounders contribute to trait variability in LCLs. While drug responses could be technically well measured on a given day, we observed significant day-to-day variability and substantial correlation to non-genetic confounders, such as baseline growth rates and metabolic state in culture. After correcting for these confounders, we were unable to detect any QTLs with genome-wide significance for drug response. A much higher proportion of variance in mRNA levels may be attributed to non-genetic factors (intra-individual variance—i.e., biological noise, levels of the EBV virus used to transform the cells, ATP levels) than to detectable eQTLs. Finally, in an attempt to improve power, we focused analysis on those genes that had both detectable eQTLs and correlation to drug response; we were unable to detect evidence that eQTL SNPs are convincingly associated with drug response in the model. While LCLs are a promising model for pharmacogenetic experiments, biological noise and in vitro artifacts may reduce power and have the potential to create spurious association due to confounding.


Nature | 2012

Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer

Elise Ruark; Katie Snape; Peter Humburg; Chey Loveday; Ilirjana Bajrami; Rachel Brough; Daniel Nava Rodrigues; Anthony Renwick; Sheila Seal; Emma Ramsay; Silvana Del Vecchio Duarte; Manuel A. Rivas; Margaret Warren-Perry; Anna Zachariou; Adriana Campion-Flora; Sandra Hanks; Anne Murray; Naser Ansari Pour; Jenny Douglas; Lorna Gregory; Andrew J. Rimmer; Neil Walker; Tsun-Po Yang; Julian Adlard; Julian Barwell; Jonathan Berg; Angela F. Brady; Carole Brewer; G Brice; Cyril Chapman

Improved sequencing technologies offer unprecedented opportunities for investigating the role of rare genetic variation in common disease. However, there are considerable challenges with respect to study design, data analysis and replication. Using pooled next-generation sequencing of 507 genes implicated in the repair of DNA in 1,150 samples, an analytical strategy focused on protein-truncating variants (PTVs) and a large-scale sequencing case–control replication experiment in 13,642 individuals, here we show that rare PTVs in the p53-inducible protein phosphatase PPM1D are associated with predisposition to breast cancer and ovarian cancer. PPM1D PTV mutations were present in 25 out of 7,781 cases versus 1 out of 5,861 controls (P = 1.12 × 10−5), including 18 mutations in 6,912 individuals with breast cancer (P = 2.42 × 10−4) and 12 mutations in 1,121 individuals with ovarian cancer (P = 3.10 × 10−9). Notably, all of the identified PPM1D PTVs were mosaic in lymphocyte DNA and clustered within a 370-base-pair region in the final exon of the gene, carboxy-terminal to the phosphatase catalytic domain. Functional studies demonstrate that the mutations result in enhanced suppression of p53 in response to ionizing radiation exposure, suggesting that the mutant alleles encode hyperactive PPM1D isoforms. Thus, although the mutations cause premature protein truncation, they do not result in the simple loss-of-function effect typically associated with this class of variant, but instead probably have a gain-of-function effect. Our results have implications for the detection and management of breast and ovarian cancer risk. More generally, these data provide new insights into the role of rare and of mosaic genetic variants in common conditions, and the use of sequencing in their identification.


Science | 2015

Human genomics. Effect of predicted protein-truncating genetic variants on the human transcriptome

Manuel A. Rivas; Matti Pirinen; Donald F. Conrad; Monkol Lek; Emily K. Tsang; Konrad J. Karczewski; Julian Maller; Kimberly R. Kukurba; David S. DeLuca; Menachem Fromer; Pedro G. Ferreira; Kevin S. Smith; Rui Zhang; Fengmei Zhao; Eric Banks; Ryan Poplin; Douglas M. Ruderfer; Shaun Purcell; Taru Tukiainen; Eric Vallabh Minikel; Peter D. Stenson; David Neil Cooper; Katharine H. Huang; Timothy J. Sullivan; Jared L. Nedzel; Carlos Bustamante; Jin Billy Li; Mark J. Daly; Roderic Guigó; Peter Donnelly

Expression, genetic variation, and tissues Human genomes show extensive genetic variation across individuals, but we have only just started documenting the effects of this variation on the regulation of gene expression. Furthermore, only a few tissues have been examined per genetic variant. In order to examine how genetic expression varies among tissues within individuals, the Genotype-Tissue Expression (GTEx) Consortium collected 1641 postmortem samples covering 54 body sites from 175 individuals. They identified quantitative genetic traits that affect gene expression and determined which of these exhibit tissue-specific expression patterns. Melé et al. measured how transcription varies among tissues, and Rivas et al. looked at how truncated protein variants affect expression across tissues. Science, this issue p. 648, p. 660, p. 666; see also p. 640 Protein-truncated variants impact gene expression levels and splicing across human tissues. [Also see Perspective by Gibson] Accurate prediction of the functional effect of genetic variation is critical for clinical genome interpretation. We systematically characterized the transcriptome effects of protein-truncating variants, a class of variants expected to have profound effects on gene function, using data from the Genotype-Tissue Expression (GTEx) and Geuvadis projects. We quantitated tissue-specific and positional effects on nonsense-mediated transcript decay and present an improved predictive model for this decay. We directly measured the effect of variants both proximal and distal to splice junctions. Furthermore, we found that robustness to heterozygous gene inactivation is not due to dosage compensation. Our results illustrate the value of transcriptome data in the functional interpretation of genetic variants.

Collaboration


Dive into the Manuel A. Rivas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge