201
|
Host RNAs, including transposons, are encapsidated by a eukaryotic single-stranded RNA virus. Proc Natl Acad Sci U S A 2012; 109:1907-12. [PMID: 22308402 DOI: 10.1073/pnas.1116168109] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Next-generation sequencing is a valuable tool in our growing understanding of the genetic diversity of viral populations. Using this technology, we have investigated the RNA content of a purified nonenveloped single-stranded RNA virus, flock house virus (FHV). We have also investigated the RNA content of virus-like particles (VLPs) of FHV and the related Nudaurelia capensis omega virus. VLPs predominantly package ribosomal RNA and transcripts of their baculoviral expression vectors. In addition, we find that 5.3% of the packaged RNAs are transposable elements derived from the Sf21 genome. This observation may be important when considering the therapeutic use of VLPs. We find that authentic FHV virions also package a variety of host RNAs, accounting for 1% of the packaged nucleic acid. Significant quantities of host messenger RNAs, ribosomal RNA, noncoding RNAs, and transposable elements are readily detected. The packaging of these host RNAs elicits the possibility of horizontal gene transfer between eukaryotic hosts that share a viral pathogen. We conclude that the genetic content of nonenveloped RNA viruses is variable, not just by genome mutation, but also in the diversity of RNA transcripts that are packaged.
Collapse
|
202
|
Armenia D, Vandenbroucke I, Fabeni L, Van Marck H, Cento V, D'Arrigo R, Van Wesenbeeck L, Scopelliti F, Micheli V, Bruzzone B, Lo Caputo S, Aerssens J, Rizzardini G, Tozzi V, Narciso P, Antinori A, Stuyver L, Perno CF, Ceccherini-Silberstein F. Study of genotypic and phenotypic HIV-1 dynamics of integrase mutations during raltegravir treatment: a refined analysis by ultra-deep 454 pyrosequencing. J Infect Dis 2012; 205:557-67. [PMID: 22238474 DOI: 10.1093/infdis/jir821] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The dynamics of raltegravir-resistant variants and their impact on virologic response in 23 HIV-1-infected patients, who started a salvage raltegravir-containing regimen, were investigated. METHODS Integrase population sequencing and Ultra-Deep-454 Pyrosequencing (UDPS) were performed on plasma samples at baseline and at raltegravir failure. All integrase mutations detected at a frequency ≥1% were considered to be reliable for the UDPS analyses. Phylogenetic and phenotypic resistance analyses were also performed. RESULTS At baseline, primary resistance mutations were not detected by both population and UDPS genotypic assays; few secondary mutations (T97A-V151I-G163R) were rarely detected and did not show any statistically association either with virologic response at 24-weeks or with the development of resistant variants at failure. At UDPS, not all resistant variants appearing early during treatment evolved as major populations during failure; only specific resistance pathways (Y143R-Q148H/R-N155H) associated with an increased rate of fitness and phenotypic resistance were selected. CONCLUSIONS Resistance to raltegravir in integrase strand transfer inhibitor-naive patients remains today a rare event, which might be changed by future extensive use of such drugs. In our study, pathways of resistance at failure were not predicted by baseline mutations, suggesting that evolution plus stochastic selection plays a major role in the appearance of integrase-resistance mutations, whereas fitness and resistance are dominant factors acting for the late selection of resistant quasispecies.
Collapse
|
203
|
Bearden A, Friedrich TC, Goldberg TL, Byrne B, Spiegel C, Schult P, Safdar N. An outbreak of the 2009 influenza a (H1N1) virus in a children's hospital. Influenza Other Respir Viruses 2012; 6:374-9. [PMID: 22212690 PMCID: PMC4941090 DOI: 10.1111/j.1750-2659.2011.00322.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Please cite this paper as: Bearden et al. (2012) An outbreak of the 2009 influenza a (H1N1) virus in a children’s hospital. Influenza and Other Respiratory Viruses 6(5), 374–379. Context Preventing nosocomial transmission of influenza is essential to reduce the morbidity and mortality associated with this infection. In October 2009, an outbreak of the 2009 influenza A (H1N1) virus occurred in a hematology ward of a children’s hospital over a 21‐day period and involved two patients and four healthcare workers. Objective To investigate nosocomial transmission of the 2009 influenza A (H1N1) virus in patients and healthcare workers. Design, setting, and participants An outbreak investigation was initiated in response to suspected nosocomial transmission of the 2009 influenza A (H1N1) virus during the peak of the 2009 pandemic. Cases were confirmed using a polymerase chain reaction (PCR) test specific for the 2009 H1N1 influenza A virus. Viruses isolated from nasopharyngeal swabs were genetically characterized using Sanger sequencing of uncloned “bulk” PCR products. Main outcome measures Virus sequencing to investigate nosocomial transmission. Results Two immunocompromised patients and four healthcare workers were found to be part of a nosocomial outbreak of the 2009 influenza A (H1N1) virus. One immunocompromised patient had a second episode of clinical influenza infection after isolation precautions had been discontinued, resulting in additional exposures. Strain‐specific PCR showed that all cases were caused by infection of the 2009 H1N1 virus. Sequencing of viral genes encoding hemagglutinin and polymerase basic subunit 2 (PB2) revealed that all viruses isolated were genetically identical at these loci, including the two episodes occurring in the same immunocompromised patient. Conclusions Prompt institution of isolation precautions is essential in preventing nosocomial outbreaks of the 2009 novel influenza A (H1N1) virus. Our data suggest that isolation precautions may need to be continued for a prolonged period of time in immunocompromised patients with influenza infection.
Collapse
Affiliation(s)
- Allison Bearden
- Sections of Infectious Diseases, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | | | | | | | | |
Collapse
|
204
|
Saeed F, Perez-Rathke A, Gwarnicki J, Berger-Wolf T, Khokhar A. High Performance Multiple Sequence Alignment System for Pyrosequencing Reads from Multiple Reference Genomes. JOURNAL OF PARALLEL AND DISTRIBUTED COMPUTING 2012; 72:83-93. [PMID: 23125479 PMCID: PMC3486434 DOI: 10.1016/j.jpdc.2011.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Genome resequencing with short reads generated from pyrosequencing generally relies on mapping the short reads against a single reference genome. However, mapping of reads from multiple reference genomes is not possible using a pairwise mapping algorithm. In order to align the reads w.r.t each other and the reference genomes, existing multiple sequence alignment(MSA) methods cannot be used because they do not take into account the position of these short reads with respect to the genome, and are highly inefficient for large number of sequences. In this paper, we develop a highly scalable parallel algorithm based on domain decomposition, referred to as P-Pyro-Align, to align such large number of reads from single or multiple reference genomes. The proposed alignment algorithm accurately aligns the erroneous reads, and has been implemented on a cluster of workstations using MPI library. Experimental results for different problem sizes are analyzed in terms of execution time, quality of the alignments, and the ability of the algorithm to handle reads from multiple haplotypes. We report high quality multiple alignment of up to 0.5 million reads. The algorithm is shown to be highly scalable and exhibits super-linear speedups with increasing number of processors.
Collapse
Affiliation(s)
- Fahad Saeed
- Department of Computer Science, University of Illinois at Chicago, IL USA
| | | | | | | | | |
Collapse
|
205
|
Tripathy S, Jiang RHY. Massively parallel sequencing technology in pathogenic microbes. Methods Mol Biol 2012; 835:271-94. [PMID: 22183660 DOI: 10.1007/978-1-61779-501-5_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Next-Generation Sequencing (NGS) methods have revolutionized various aspects of genomics including transcriptome analysis. Digital expression analysis is all set to replace analog expression analysis that uses microarray chips through their cost-effectiveness, reproducibility, accuracy, and speed. The last 2 years have seen a surge in the development of statistical methods and software tools for analysis and visualization of NGS data. Large amounts of NGS data are available for pathogenic fungi and oomycetes. As the analysis results start pouring in, it brings about a paradigm shift in the understanding of host pathogen interactions with discovery of new transcripts, splice variants, mutations, regulatory elements, and epigenetic controls. Here we describe the core technology of the new sequencing platforms, the methodology of data analysis, and different aspects of applications.
Collapse
Affiliation(s)
- Sucheta Tripathy
- Virginia Bioinformatics Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| | | |
Collapse
|
206
|
García-González C, García-Bujalance S, Ruiz-Carrascoso G, Arribas JR, González-García J, Bernardino JI, Pascual-Pareja JF, Martínez-Prats L, Delgado R, Mingorance J. Detection and quantification of the K103N mutation in HIV reverse transcriptase by pyrosequencing. Diagn Microbiol Infect Dis 2012; 72:90-6. [DOI: 10.1016/j.diagmicrobio.2011.09.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 09/14/2011] [Accepted: 09/22/2011] [Indexed: 11/16/2022]
|
207
|
High-throughput VDJ sequencing for quantification of minimal residual disease in chronic lymphocytic leukemia and immune reconstitution assessment. Proc Natl Acad Sci U S A 2011; 108:21194-9. [PMID: 22160699 DOI: 10.1073/pnas.1118357109] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The primary cause of poor outcome following allogeneic hematopoietic cell transplantation (HCT) for chronic lymphocytic leukemia (CLL) is disease recurrence. Detection of increasing minimal residual disease (MRD) following HCT may permit early intervention to prevent clinical relapse; however, MRD quantification remains an uncommon diagnostic test because of logistical and financial barriers to widespread use. Here we describe a method for quantifying CLL MRD using widely available consensus primers for amplification of all Ig heavy chain (IGH) genes in a mixture of peripheral blood mononuclear cells, followed by high-throughput sequencing (HTS) for disease-specific IGH sequence quantification. To achieve accurate MRD quantification, we developed a systematic bioinformatic methodology to aggregate cancer clone sequence variants arising from systematic and random artifacts occurring during IGH-HTS. We then compared the sensitivity of IGH-HTS, flow cytometry, and allele-specific oligonucleotide PCR for MRD quantification in 28 samples collected from 6 CLL patients following allogeneic HCT. Using amplimer libraries generated with consensus primers from patient blood samples, we demonstrate the sensitivity of IGH-HTS with 454 pyrosequencing to be 10(-5), with a high correlation between quantification by allele-specific oligonucleotide PCR and IGH-HTS (r = 0.85). From the same dataset used to quantify MRD, IGH-HTS also allowed us to profile IGH repertoire reconstitution after HCT-information not provided by the other MRD methods. IGH-HTS using consensus primers will broaden the availability of MRD quantification in CLL and other B cell malignancies, and this approach has potential for quantitative evaluation of immune diversification following transplant and nontransplant therapies.
Collapse
|
208
|
Accurate sampling and deep sequencing of the HIV-1 protease gene using a Primer ID. Proc Natl Acad Sci U S A 2011; 108:20166-71. [PMID: 22135472 DOI: 10.1073/pnas.1110064108] [Citation(s) in RCA: 301] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Viruses can create complex genetic populations within a host, and deep sequencing technologies allow extensive sampling of these populations. Limitations of these technologies, however, potentially bias this sampling, particularly when a PCR step precedes the sequencing protocol. Typically, an unknown number of templates are used in initiating the PCR amplification, and this can lead to unrecognized sequence resampling creating apparent homogeneity; also, PCR-mediated recombination can disrupt linkage, and differential amplification can skew allele frequency. Finally, misincorporation of nucleotides during PCR and errors during the sequencing protocol can inflate diversity. We have solved these problems by including a random sequence tag in the initial primer such that each template receives a unique Primer ID. After sequencing, repeated identification of a Primer ID reveals sequence resampling. These resampled sequences are then used to create an accurate consensus sequence for each template, correcting for recombination, allelic skewing, and misincorporation/sequencing errors. The resulting population of consensus sequences directly represents the initial sampled templates. We applied this approach to the HIV-1 protease (pro) gene to view the distribution of sequence variation of a complex viral population within a host. We identified major and minor polymorphisms at coding and noncoding positions. In addition, we observed dynamic genetic changes within the population during intermittent drug exposure, including the emergence of multiple resistant alleles. These results provide an unprecedented view of a complex viral population in the absence of PCR resampling.
Collapse
|
209
|
van Westen GJP, Wegner JK, Geluykens P, Kwanten L, Vereycken I, Peeters A, IJzerman AP, van Vlijmen HWT, Bender A. Which compound to select in lead optimization? Prospectively validated proteochemometric models guide preclinical development. PLoS One 2011; 6:e27518. [PMID: 22132107 PMCID: PMC3223189 DOI: 10.1371/journal.pone.0027518] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 10/18/2011] [Indexed: 11/19/2022] Open
Abstract
In quite a few diseases, drug resistance due to target variability poses a serious problem in pharmacotherapy. This is certainly true for HIV, and hence, it is often unknown which drug is best to use or to develop against an individual HIV strain. In this work we applied 'proteochemometric' modeling of HIV Non-Nucleoside Reverse Transcriptase (NNRTI) inhibitors to support preclinical development by predicting compound performance on multiple mutants in the lead selection stage. Proteochemometric models are based on both small molecule and target properties and can thus capture multi-target activity relationships simultaneously, the targets in this case being a set of 14 HIV Reverse Transcriptase (RT) mutants. We validated our model by experimentally confirming model predictions for 317 untested compound-mutant pairs, with a prediction error comparable with assay variability (RMSE 0.62). Furthermore, dependent on the similarity of a new mutant to the training set, we could predict with high accuracy which compound will be most effective on a sequence with a previously unknown genotype. Hence, our models allow the evaluation of compound performance on untested sequences and the selection of the most promising leads for further preclinical research. The modeling concept is likely to be applicable also to other target families with genetic variability like other viruses or bacteria, or with similar orthologs like GPCRs.
Collapse
Affiliation(s)
- Gerard J. P. van Westen
- Division of Medicinal Chemistry, Leiden/Amsterdam Center for Drug Research, Leiden, The Netherlands
| | | | | | | | | | | | - Adriaan P. IJzerman
- Division of Medicinal Chemistry, Leiden/Amsterdam Center for Drug Research, Leiden, The Netherlands
| | - Herman W. T. van Vlijmen
- Division of Medicinal Chemistry, Leiden/Amsterdam Center for Drug Research, Leiden, The Netherlands
- Tibotec BVBA, Beerse, Belgium
| | - Andreas Bender
- Division of Medicinal Chemistry, Leiden/Amsterdam Center for Drug Research, Leiden, The Netherlands
- Unilever Centre for Molecular Science Informatics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
210
|
Brooks MJ, Rajasimha HK, Roger JE, Swaroop A. Next-generation sequencing facilitates quantitative analysis of wild-type and Nrl(-/-) retinal transcriptomes. Mol Vis 2011; 17:3034-54. [PMID: 22162623 PMCID: PMC3233386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 11/18/2011] [Indexed: 11/08/2022] Open
Abstract
PURPOSE Next-generation sequencing (NGS) has revolutionized systems-based analysis of cellular pathways. The goals of this study are to compare NGS-derived retinal transcriptome profiling (RNA-seq) to microarray and quantitative reverse transcription polymerase chain reaction (qRT-PCR) methods and to evaluate protocols for optimal high-throughput data analysis. METHODS Retinal mRNA profiles of 21-day-old wild-type (WT) and neural retina leucine zipper knockout (Nrl(-/-)) mice were generated by deep sequencing, in triplicate, using Illumina GAIIx. The sequence reads that passed quality filters were analyzed at the transcript isoform level with two methods: Burrows-Wheeler Aligner (BWA) followed by ANOVA (ANOVA) and TopHat followed by Cufflinks. qRT-PCR validation was performed using TaqMan and SYBR Green assays. RESULTS Using an optimized data analysis workflow, we mapped about 30 million sequence reads per sample to the mouse genome (build mm9) and identified 16,014 transcripts in the retinas of WT and Nrl(-/-) mice with BWA workflow and 34,115 transcripts with TopHat workflow. RNA-seq data confirmed stable expression of 25 known housekeeping genes, and 12 of these were validated with qRT-PCR. RNA-seq data had a linear relationship with qRT-PCR for more than four orders of magnitude and a goodness of fit (R(2)) of 0.8798. Approximately 10% of the transcripts showed differential expression between the WT and Nrl(-/-) retina, with a fold change ≥1.5 and p value <0.05. Altered expression of 25 genes was confirmed with qRT-PCR, demonstrating the high degree of sensitivity of the RNA-seq method. Hierarchical clustering of differentially expressed genes uncovered several as yet uncharacterized genes that may contribute to retinal function. Data analysis with BWA and TopHat workflows revealed a significant overlap yet provided complementary insights in transcriptome profiling. CONCLUSIONS Our study represents the first detailed analysis of retinal transcriptomes, with biologic replicates, generated by RNA-seq technology. The optimized data analysis workflows reported here should provide a framework for comparative investigations of expression profiles. Our results show that NGS offers a comprehensive and more accurate quantitative and qualitative evaluation of mRNA content within a cell or tissue. We conclude that RNA-seq based transcriptome characterization would expedite genetic network analyses and permit the dissection of complex biologic functions.
Collapse
|
211
|
Prosperi MCF, Salemi M. QuRe: software for viral quasispecies reconstruction from next-generation sequencing data. ACTA ACUST UNITED AC 2011; 28:132-3. [PMID: 22088846 DOI: 10.1093/bioinformatics/btr627] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
SUMMARY Next-generation sequencing (NGS) is an ideal framework for the characterization of highly variable pathogens, with a deep resolution able to capture minority variants. However, the reconstruction of all variants of a viral population infecting a host is a challenging task for genome regions larger than the average NGS read length. QuRe is a program for viral quasispecies reconstruction, specifically developed to analyze long read (>100 bp) NGS data. The software performs alignments of sequence fragments against a reference genome, finds an optimal division of the genome into sliding windows based on coverage and diversity and attempts to reconstruct all the individual sequences of the viral quasispecies--along with their prevalence--using a heuristic algorithm, which matches multinomial distributions of distinct viral variants overlapping across the genome division. QuRe comes with a built-in Poisson error correction method and a post-reconstruction probabilistic clustering, both parameterized on given error rates in homopolymeric and non-homopolymeric regions. AVAILABILITY QuRe is platform-independent, multi-threaded software implemented in Java. It is distributed under the GNU General Public License, available at https://sourceforge.net/projects/qure/. CONTACT ahnven@yahoo.it; ahnven@gmail.com SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mattia C F Prosperi
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610-3633, USA
| | | |
Collapse
|
212
|
Applications of next-generation sequencing technologies to diagnostic virology. Int J Mol Sci 2011; 12:7861-84. [PMID: 22174638 PMCID: PMC3233444 DOI: 10.3390/ijms12117861] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 10/20/2011] [Accepted: 11/07/2011] [Indexed: 02/07/2023] Open
Abstract
Novel DNA sequencing techniques, referred to as “next-generation” sequencing (NGS), provide high speed and throughput that can produce an enormous volume of sequences with many possible applications in research and diagnostic settings. In this article, we provide an overview of the many applications of NGS in diagnostic virology. NGS techniques have been used for high-throughput whole viral genome sequencing, such as sequencing of new influenza viruses, for detection of viral genome variability and evolution within the host, such as investigation of human immunodeficiency virus and human hepatitis C virus quasispecies, and monitoring of low-abundance antiviral drug-resistance mutations. NGS techniques have been applied to metagenomics-based strategies for the detection of unexpected disease-associated viruses and for the discovery of novel human viruses, including cancer-related viruses. Finally, the human virome in healthy and disease conditions has been described by NGS-based metagenomics.
Collapse
|
213
|
Rosani U, Varotto L, Rossi A, Roch P, Novoa B, Figueras A, Pallavicini A, Venier P. Massively parallel amplicon sequencing reveals isotype-specific variability of antimicrobial peptide transcripts in Mytilus galloprovincialis. PLoS One 2011; 6:e26680. [PMID: 22087233 PMCID: PMC3210125 DOI: 10.1371/journal.pone.0026680] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Accepted: 10/02/2011] [Indexed: 11/19/2022] Open
Abstract
Background Effective innate responses against potential pathogens are essential in the living world and possibly contributed to the evolutionary success of invertebrates. Taken together, antimicrobial peptide (AMP) precursors of defensin, mytilin, myticin and mytimycin can represent about 40% of the hemocyte transcriptome in mussels injected with viral-like and bacterial preparations, and unique profiles of myticin C variants are expressed in single mussels. Based on amplicon pyrosequencing, we have ascertained and compared the natural and Vibrio-induced diversity of AMP transcripts in mussel hemocytes from three European regions. Methodology/Principal Findings Hemolymph was collected from mussels farmed in the coastal regions of Palavas (France), Vigo (Spain) and Venice (Italy). To represent the AMP families known in M. galloprovincialis, nine transcript sequences have been selected, amplified from hemocyte RNA and subjected to pyrosequencing. Hemolymph from farmed (offshore) and wild (lagoon) Venice mussels, both injected with 107Vibrio cells, were similarly processed. Amplicon pyrosequencing emphasized the AMP transcript diversity, with Single Nucleotide Changes (SNC) minimal for mytilin B/C and maximal for arthropod-like defensin and myticin C. Ratio of non-synonymous vs. synonymous changes also greatly differed between AMP isotypes. Overall, each amplicon revealed similar levels of nucleotidic variation across geographical regions, with two main sequence patterns confirmed for mytimycin and no substantial changes after immunostimulation. Conclusions/Significance Barcoding and bidirectional pyrosequencing allowed us to map and compare the transcript diversity of known mussel AMPs. Though most of the genuine cds variation was common to the analyzed samples we could estimate from 9 to 106 peptide variants in hemolymph pools representing 100 mussels, depending on the AMP isoform and sampling site. In this study, no prevailing SNC patterns related to geographical origin or Vibrio injection emerged. Whether or not the contact with potential pathogens can increase the amount of AMP transcript variants in mussels requires additional study.
Collapse
Affiliation(s)
- Umberto Rosani
- Department of Biology, University of Padua, Padova, Italy
- * E-mail: (UR); (PV)
| | - Laura Varotto
- Department of Biology, University of Padua, Padova, Italy
| | - Alberta Rossi
- Department of Biology, University of Padua, Padova, Italy
| | - Philippe Roch
- Ecologie des Systèmes Marins et Côtiers, CNRS-IRD-University of Montpellier 2, Montpellier, France
| | - Beatriz Novoa
- Instituto de Investigaciones Marinas, CSIC, Vigo, Spain
| | | | | | - Paola Venier
- Department of Biology, University of Padua, Padova, Italy
- * E-mail: (UR); (PV)
| |
Collapse
|
214
|
Altmann A, Weber P, Quast C, Rex-Haffner M, Binder EB, Müller-Myhsok B. vipR: variant identification in pooled DNA using R. Bioinformatics 2011; 27:i77-84. [PMID: 21685105 PMCID: PMC3117388 DOI: 10.1093/bioinformatics/btr205] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Motivation: High-throughput-sequencing (HTS) technologies are the method of choice for screening the human genome for rare sequence variants causing susceptibility to complex diseases. Unfortunately, preparation of samples for a large number of individuals is still very cost- and labor intensive. Thus, recently, screens for rare sequence variants were carried out in samples of pooled DNA, in which equimolar amounts of DNA from multiple individuals are mixed prior to sequencing with HTS. The resulting sequence data, however, poses a bioinformatics challenge: the discrimination of sequencing errors from real sequence variants present at a low frequency in the DNA pool. Results: Our method vipR uses data from multiple DNA pools in order to compensate for differences in sequencing error rates along the sequenced region. More precisely, instead of aiming at discriminating sequence variants from sequencing errors, vipR identifies sequence positions that exhibit significantly different minor allele frequencies in at least two DNA pools using the Skellam distribution. The performance of vipR was compared with three other models on data from a targeted resequencing study of the TMEM132D locus in 600 individuals distributed over four DNA pools. Performance of the methods was computed on SNPs that were also genotyped individually using a MALDI-TOF technique. On a set of 82 sequence variants, vipR achieved an average sensitivity of 0.80 at an average specificity of 0.92, thus outperforming the reference methods by at least 0.17 in specificity at comparable sensitivity. Availability: The code of vipR is freely available via: http://sourceforge.net/projects/htsvipr/ Contact:altmann@mpipsykl.mpg.de
Collapse
Affiliation(s)
- Andre Altmann
- Department of Statistical Genetics, Max Planck Institute of Psychiatry, Munich, Germany.
| | | | | | | | | | | |
Collapse
|
215
|
Abstract
OBJECTIVE Our objective was to analyze the pathways leading to resistance of HIV to the integrase (IN) inhibitor raltegravir (RAL). DESIGN Three HIV-infected individuals exhibiting RAL resistance pathway switching were characterized using longitudinal analysis of viral samples from plasma. METHODS 454/Roche pyrosequencing was used to generate approximately 74,000 sequence reads from the integrase coding region. Effects of error were controlled by denoising with Pyronoise, and by comparison to approximately 142,000 control reads from HIV(NL4-3). Viral lineages were modeled quantitatively using viral serial pathway analysis (vSPA). RESULTS All three patients showed transitions from the N155H pathway to the Q148H/G140S pathway. Analysis with vSPA revealed complex pathways to the final genotype, probably involving both de-novo mutation and recombination. No reads contained both the N155H and Q148H drug resistance mutations (DRMs), indicating that the double mutant is not a prominent intermediate, consistent with low fitness. To characterize possible drug-resistant variants circulating prior to therapy, we sequenced approximately 70,000 reads from samples collected prior to initiating treatment. Although some preexisting drug-resistant variants were detected, N155H, the first major DRM present after initiating RAL therapy, was not detected. CONCLUSION The main DRMs are present at very low levels if at all prior to initiating therapy. We also outline general methods for deep sequence analysis of DRMs in longitudinal HIV samples.
Collapse
|
216
|
Liang B, Luo M, Scott-Herridge J, Semeniuk C, Mendoza M, Capina R, Sheardown B, Ji H, Kimani J, Ball BT, Van Domselaar G, Graham M, Tyler S, Jones SJM, Plummer FA. A comparison of parallel pyrosequencing and sanger clone-based sequencing and its impact on the characterization of the genetic diversity of HIV-1. PLoS One 2011; 6:e26745. [PMID: 22039546 PMCID: PMC3198814 DOI: 10.1371/journal.pone.0026745] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 10/03/2011] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Pyrosequencing technology has the potential to rapidly sequence HIV-1 viral quasispecies without requiring the traditional approach of cloning. In this study, we investigated the utility of ultra-deep pyrosequencing to characterize genetic diversity of the HIV-1 gag quasispecies and assessed the possible contribution of pyrosequencing technology in studying HIV-1 biology and evolution. METHODOLOGY/PRINCIPAL FINDINGS HIV-1 gag gene was amplified from 96 patients using nested PCR. The PCR products were cloned and sequenced using capillary based Sanger fluorescent dideoxy termination sequencing. The same PCR products were also directly sequenced using the 454 pyrosequencing technology. The two sequencing methods were evaluated for their ability to characterize quasispecies variation, and to reveal sites under host immune pressure for their putative functional significance. A total of 14,034 variations were identified by 454 pyrosequencing versus 3,632 variations by Sanger clone-based (SCB) sequencing. 11,050 of these variations were detected only by pyrosequencing. These undetected variations were located in the HIV-1 Gag region which is known to contain putative cytotoxic T lymphocyte (CTL) and neutralizing antibody epitopes, and sites related to virus assembly and packaging. Analysis of the positively selected sites derived by the two sequencing methods identified several differences. All of them were located within the CTL epitope regions. CONCLUSIONS/SIGNIFICANCE Ultra-deep pyrosequencing has proven to be a powerful tool for characterization of HIV-1 genetic diversity with enhanced sensitivity, efficiency, and accuracy. It also improved reliability of downstream evolutionary and functional analysis of HIV-1 quasispecies.
Collapse
Affiliation(s)
- Binhua Liang
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Flaherty P, Natsoulis G, Muralidharan O, Winters M, Buenrostro J, Bell J, Brown S, Holodniy M, Zhang N, Ji HP. Ultrasensitive detection of rare mutations using next-generation targeted resequencing. Nucleic Acids Res 2011; 40:e2. [PMID: 22013163 PMCID: PMC3245950 DOI: 10.1093/nar/gkr861] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
With next-generation DNA sequencing technologies, one can interrogate a specific genomic region of interest at very high depth of coverage and identify less prevalent, rare mutations in heterogeneous clinical samples. However, the mutation detection levels are limited by the error rate of the sequencing technology as well as by the availability of variant-calling algorithms with high statistical power and low false positive rates. We demonstrate that we can robustly detect mutations at 0.1% fractional representation. This represents accurate detection of one mutant per every 1000 wild-type alleles. To achieve this sensitive level of mutation detection, we integrate a high accuracy indexing strategy and reference replication for estimating sequencing error variance. We employ a statistical model to estimate the error rate at each position of the reference and to quantify the fraction of variant base in the sample. Our method is highly specific (99%) and sensitive (100%) when applied to a known 0.1% sample fraction admixture of two synthetic DNA samples to validate our method. As a clinical application of this method, we analyzed nine clinical samples of H1N1 influenza A and detected an oseltamivir (antiviral therapy) resistance mutation in the H1N1 neuraminidase gene at a sample fraction of 0.18%.
Collapse
Affiliation(s)
- Patrick Flaherty
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA 94304, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Homs M, Buti M, Quer J, Jardí R, Schaper M, Tabernero D, Ortega I, Sanchez A, Esteban R, Rodriguez-Frias F. Ultra-deep pyrosequencing analysis of the hepatitis B virus preCore region and main catalytic motif of the viral polymerase in the same viral genome. Nucleic Acids Res 2011; 39:8457-8471. [PMID: 21742757 PMCID: PMC3201856 DOI: 10.1093/nar/gkr451] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 04/27/2011] [Accepted: 05/16/2011] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) pregenomic RNA contains a hairpin structure (ε) located in the preCore region, essential for viral replication. ε stability is enhanced by the presence of preCore variants and ε is recognized by the HBV polymerase (Pol). Mutations in the retrotranscriptase domain (YMDD) of Pol are associated with treatment resistance. The aim of this study was to analyze the preCore region and YMDD motif by ultra-deep pyrosequencing (UDPS). To evaluate the UDPS error rate, an internal control sequence was inserted in the amplicon. A newly developed technique enabled simultaneous analysis of the preCore region and Pol in the same viral genome, as well as the conserved sequence of the internal control. Nucleotide errors in HindIII yielded a UDPS error rate <0.05%. UDPS study confirmed the possibility of simultaneous detection of preCore and YMDD mutations, and demonstrated the complexity of the HBV quasispecies and cooperation between viruses. Thermodynamic stability of the ε signal was found to be the main constraint for selecting main preCore mutations. Analysis of ε-signal variability suggested the essential nature of the ε structural motif and that certain nucleotides may be involved in ε signal functions.
Collapse
Affiliation(s)
- Maria Homs
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| | - Maria Buti
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| | - Josep Quer
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| | - Rosendo Jardí
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| | - Melanie Schaper
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| | - David Tabernero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| | - Israel Ortega
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| | - Alex Sanchez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| | - Rafael Esteban
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| | - Francisco Rodriguez-Frias
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Carlos III Corsega 180, 08036, Barcelona, Department of Biochemistry, Department of Hepatology, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona Passeig Vall d'Hebron 119–129, 08035, Barcelona and Statistics and Bioinformatics Unit, Research Institut, Hospital Vall d’Hebron Passeig Vall d'Hebron 119–129, 08035, Barcelona, Spain
| |
Collapse
|
219
|
Gabriel C, Stabentheiner S, Danzer M, Pröll J. What Next? The Next Transit from Biology to Diagnostics: Next Generation Sequencing for Immunogenetics. ACTA ACUST UNITED AC 2011; 38:308-317. [PMID: 22670120 DOI: 10.1159/000332433] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 09/01/2011] [Indexed: 01/06/2023]
Abstract
The human genome project triggered the introduction of next generation sequencing (NGS) systems. Although originally developed for total genome sequencing, metagenomics and plant genetics, the ultra-deep sequencing feature of NGS was utilized for diagnostic purposes in HIV resistance and tropism as well in detecting new mutations and tumor clones in oncology. Recent publications exploited the feature of clonal sequencing for immunogenetics to dissolve the growing number of ambiguities. This concept is quite reliable if all exons of interest are tested and the amplification region includes flanking introns. Challenging questions on quality control, cost effectiveness, workflow, and management of enormous loads of data remain if NGS is considered as routine method in the immunogenetics laboratory. If solved, NGS has big potential to have a major impact on immunogenetics by way of providing ambiguity-free HLA-typing results faster, but will also have a great influence on how immunogenetics testing and workflows are organized.
Collapse
|
220
|
Ramachandran S, Zhai X, Thai H, Campo DS, Xia G, Ganova-Raeva LM, Drobeniuc J, Khudyakov YE. Evaluation of intra-host variants of the entire hepatitis B virus genome. PLoS One 2011; 6:e25232. [PMID: 21949887 PMCID: PMC3176825 DOI: 10.1371/journal.pone.0025232] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 08/30/2011] [Indexed: 02/07/2023] Open
Abstract
Genetic analysis of hepatitis B virus (HBV) frequently involves study of intra-host variants, identification of which is commonly achieved using short regions of the HBV genome. However, the use of short sequences significantly limits evaluation of genetic relatedness among HBV strains. Although analysis of HBV complete genomes using genetic cloning has been developed, its application is highly labor intensive and practiced only infrequently. We describe here a novel approach to whole genome (WG) HBV quasispecies analysis based on end-point, limiting-dilution real-time PCR (EPLD-PCR) for amplification of single HBV genome variants, and their subsequent sequencing. EPLD-PCR was used to analyze WG quasispecies from serum samples of patients (n = 38) infected with HBV genotypes A, B, C, D, E and G. Phylogenetic analysis of the EPLD-isolated HBV-WG quasispecies showed the presence of mixed genotypes, recombinant variants and sub-populations of the virus. A critical observation was that HBV-WG consensus sequences obtained by direct sequencing of PCR fragments without EPLD are genetically close, but not always identical to the major HBV variants in the intra-host population, thus indicating that consensus sequences should be judiciously used in genetic analysis. Sequence-based studies of HBV WG quasispecies should afford a more accurate assessment of HBV evolution in various clinical and epidemiological settings.
Collapse
Affiliation(s)
- Sumathi Ramachandran
- Molecular Epidemiology and Bioinformatics Laboratory, Division of Viral Hepatitis, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
221
|
High levels of genetic variation within core Helicoverpa armigera nucleopolyhedrovirus genes. Virus Genes 2011; 44:149-62. [DOI: 10.1007/s11262-011-0660-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 08/08/2011] [Indexed: 12/11/2022]
|
222
|
Alteri C, Santoro MM, Abbate I, Rozera G, Bruselles A, Bartolini B, Gori C, Forbici F, Orchi N, Tozzi V, Palamara G, Antinori A, Narciso P, Girardi E, Svicher V, Ceccherini-Silberstein F, Capobianchi MR, Perno CF. 'Sentinel' mutations in standard population sequencing can predict the presence of HIV-1 reverse transcriptase major mutations detectable only by ultra-deep pyrosequencing. J Antimicrob Chemother 2011; 66:2615-23. [DOI: 10.1093/jac/dkr354] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
223
|
Sequential bottlenecks drive viral evolution in early acute hepatitis C virus infection. PLoS Pathog 2011; 7:e1002243. [PMID: 21912520 PMCID: PMC3164670 DOI: 10.1371/journal.ppat.1002243] [Citation(s) in RCA: 185] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 07/12/2011] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C is a pandemic human RNA virus, which commonly causes chronic infection and liver disease. The characterization of viral populations that successfully initiate infection, and also those that drive progression to chronicity is instrumental for understanding pathogenesis and vaccine design. A comprehensive and longitudinal analysis of the viral population was conducted in four subjects followed from very early acute infection to resolution of disease outcome. By means of next generation sequencing (NGS) and standard cloning/Sanger sequencing, genetic diversity and viral variants were quantified over the course of the infection at frequencies as low as 0.1%. Phylogenetic analysis of reassembled viral variants revealed acute infection was dominated by two sequential bottleneck events, irrespective of subsequent chronicity or clearance. The first bottleneck was associated with transmission, with one to two viral variants successfully establishing infection. The second occurred approximately 100 days post-infection, and was characterized by a decline in viral diversity. In the two subjects who developed chronic infection, this second bottleneck was followed by the emergence of a new viral population, which evolved from the founder variants via a selective sweep with fixation in a small number of mutated sites. The diversity at sites with non-synonymous mutation was higher in predicted cytotoxic T cell epitopes, suggesting immune-driven evolution. These results provide the first detailed analysis of early within-host evolution of HCV, indicating strong selective forces limit viral evolution in the acute phase of infection. Primary hepatitis C (HCV) infection is typically asymptomatic and commonly results in persistent infection. The characteristics of early infection remain undefined. Four subjects were studied longitudinally from within a few weeks of transmission until resolution of outcome, via a full genome analysis of viral evolution. In the acute phase (<100 days post-infection) there were two periods with a major reduction in genetic diversity (i.e. a bottleneck) irrespective of subsequent clearance (n = 2) or chronic infection (n = 2). The first bottleneck was associated with transmission, with generally only one ‘founder’ virus successfully establishing infection. The second occurred following the primary peak in viraemia, concomitant with seroconversion, approximately 100 days post-infection. In the subjects who became chronically infected, the second bottleneck was followed by emergence of a new cluster of variants, which evolved from the founder(s), and carried only a small number of mutated residues that reached fixation. Some fixations occurred in known targets of CD8 cytotoxic T cell and neutralizing antibody responses. These results indicate a common evolutionary pattern, independent of disease outcome in the acute phase of HCV infection, with strong signatures of selective pressures driving the transition into chronic infection. These novel data will inform preventative vaccine strategies.
Collapse
|
224
|
Minor variant detection in amplicons using 454 massive parallel pyrosequencing: experiences and considerations for successful applications. Biotechniques 2011; 51:167-77. [DOI: 10.2144/000113733] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 08/09/2011] [Indexed: 11/23/2022] Open
Abstract
Ultra-deep sequencing (UDS) of amplicons is a major application for next-generation sequencing tech nologies, even more so for the 454 Genome Sequencer FLX. Especially for this application, errors that might be introduced during any of the sample processing or data analysis steps should be avoided or at least recognized, as they might lead to aberrant sequence variant calling. Since 454 pyrosequencing relies on PCR-driven target amplification, it is key to differentiate errors introduced during the amplification step from genuine minority variants. Thereto, optimal primer design is imperative because primer selection, primer dimer formation, and nonspecific binding may all affect the quality and outcome of amplicon-based deep sequencing. Also, other intrinsic PCR characteristics including amplification drift and the formation of secondary structures may influence sequencing data quality. We illustrate these phenomena using real life case studies and propose experimental and analytical evidence-based solutions for effective practice. Furthermore, because accuracy of the DNA polymerase is vital for reliable UDS results, a comparative analysis of error profiles from seven different DNA polymerases was performed and experimentally as-sessed in parallel by 454 sequencing. Finally, intra and interrun variability evaluation of the 454 sequencing protocol revealed highly reproducible results in amplicon-based UDS.
Collapse
|
225
|
Efficient identification of human immunodeficiency virus type 1 mutants resistant to a protease inhibitor by using a random mutant library. Antimicrob Agents Chemother 2011; 55:5090-8. [PMID: 21876045 DOI: 10.1128/aac.00687-11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Emergence of drug-resistant mutant viruses during the course of antiretroviral therapy is a major hurdle that limits the success of chemotherapeutic treatment to suppress human immunodeficiency virus type 1 (HIV-1) replication and AIDS progression. Development of new drugs and careful patient management based on resistance genotyping data are important for enhancing therapeutic efficacy. However, identifying changes leading to drug resistance can take years of clinical studies, and conventional in vitro assays are limited in generating reliable drug resistance data. Here we present an efficient in vitro screening assay for selecting drug-resistant variants from a library of randomly mutated HIV-1 strains generated by transposon-directed base-exchange mutagenesis. As a test of principle, we screened a library of mutant HIV-1 strains containing random mutations in the protease gene by using a reporter T-cell line in the presence of the protease inhibitor (PI) nelfinavir (NFV). Analysis of replicating viruses from a single round of infection identified 50 amino acid substitutions at 35 HIV-1 protease residue positions. The selected mutant viruses showed specific resistance to NFV and included most of the known NFV resistance mutations. Therefore, the new assay is efficient for identifying changes leading to drug resistance. The data also provide insights into the molecular mechanisms underlying the development of drug resistance.
Collapse
|
226
|
Stelzl E, Pröll J, Bizon B, Niklas N, Danzer M, Hackl C, Stabentheiner S, Gabriel C, Kessler HH. Human immunodeficiency virus type 1 drug resistance testing: Evaluation of a new ultra-deep sequencing-based protocol and comparison with the TRUGENE HIV-1 Genotyping Kit. J Virol Methods 2011; 178:94-7. [PMID: 21907239 DOI: 10.1016/j.jviromet.2011.08.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 08/19/2011] [Accepted: 08/22/2011] [Indexed: 12/27/2022]
Abstract
Genotypic HIV-1 drug resistance testing with standard Sanger sequencing is limited to the detection of mutations with >20% prevalence. A new protocol for variant detection of protease and reverse transcriptase genes of HIV-1 genotype B samples with ultra-deep sequencing on the GS-FLX sequencer (Roche 454 Life Sciences, Branford, CT) was evaluated. The new technology was compared with the standard Sanger sequencing method. For accuracy testing, genotype B samples obtained from proficiency panels were examined with ultra-deep sequencing. Reproducibility was determined by repeat GS-FLX sequencing of 21 clinical samples. Clinical performance was evaluated with 44 samples and the results were compared to the TRUGENE HIV-1 Genotyping Kit (Siemens Healthcare Diagnostics, Tarrytown, NY). Sequences generated with both protocols were analyzed using the Stanford University HIV drug resistance database. When accuracy was tested, 316 of 317 mutation codons included in the analysis of proficiency panels could be identified correctly with ultra-deep sequencing. Reproducibility testing resulted in a correlation value of R(2)=0.969. Analysis of 44 routine clinical samples with the Stanford University HIV drug resistance database revealed a total number of 269 and 171 mutations by the ultra-deep and standard Sanger sequencing, respectively. Drug resistance interpretations showed differences for 11 samples. With ultra-deep sequencing, total time to result was four times longer in comparison to standard Sanger sequencing. Manual work was increased significantly using the new protocol. The ultra-deep sequencing protocol showed good accuracy and reproducibility. However, automation and shorter time to obtain results are essential for use in the routine diagnostic laboratory.
Collapse
Affiliation(s)
- Evelyn Stelzl
- Research Unit Molecular Diagnostics and Molecular Diagnostics Laboratory, IHMEM, Medical University of Graz, Graz, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Beerenwinkel N, Zagordi O. Ultra-deep sequencing for the analysis of viral populations. Curr Opin Virol 2011; 1:413-8. [PMID: 22440844 DOI: 10.1016/j.coviro.2011.07.008] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 07/22/2011] [Indexed: 12/17/2022]
Abstract
Next-generation sequencing allows for cost-effective probing of virus populations at an unprecedented level of detail. The massively parallel sequencing approach can detect low-frequency mutations and it provides a snapshot of the entire virus population. However, analyzing ultra-deep sequencing data obtained from diverse virus populations is challenging because of PCR and sequencing errors and short read lengths, such that the experiment provides only indirect evidence of the underlying viral population structure. Recent computational and statistical advances allow for accommodating some of the confounding factors, including methods for read error correction, haplotype reconstruction, and haplotype frequency estimation. With these methods ultra-deep sequencing can be more reliably used to analyze, in a quantitative manner, the genetic diversity of virus populations.
Collapse
Affiliation(s)
- Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
| | | |
Collapse
|
228
|
Hunt GM, Coovadia A, Abrams EJ, Sherman G, Meyers T, Morris L, Kuhn L. HIV-1 drug resistance at antiretroviral treatment initiation in children previously exposed to single-dose nevirapine. AIDS 2011; 25:1461-9. [PMID: 21633285 PMCID: PMC4547386 DOI: 10.1097/qad.0b013e3283492180] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To describe the prevalence of HIV-1 drug resistance mutations at the time of treatment initiation in a large cohort of HIV-infected children previously exposed to single-dose nevirapine (sdNVP) for prevention of transmission. DESIGN Drug resistance mutations were measured pretreatment in 255 infants and young children under 2 years of age in South Africa exposed to sdNVP and initiating ritonavir-boosted lopinavir-based therapy. Those who achieved viral suppression were randomized to either continue the primary regimen or to switch to a nevirapine-based regimen. Pretreatment samples were tested using population sequencing and real time allele-specific PCR (AS-PCR) to detect Y181C and K103N minority variants. Those with confirmed viremia more than 1000 copies/ml by 52 weeks postrandomization in the switch group were defined as having viral failure. RESULTS Nonnucleoside reverse transcriptase inhibitor (NNRTI) mutations, predominantly Y181C, were detected by either method in 62% of infants less than 6 months of age, in 39% of children 6-12 months of age, 22% 12-18 months, and 16% 18-24 months (P = <0.0001). NNRTI mutations detected by genotyping, but not K103N or Y181C mutations detected only by AS-PCR, were associated with viral failure in the switch group. CONCLUSION The prevalence of mutations known to compromise primary NNRTI-based therapy is high in sdNVP-exposed children, supporting current guidelines recommending use of protease inhibitor-based regimens for young children. Standard genotyping is adequate to identify children who could benefit from switching to NNRTI-based therapy.
Collapse
Affiliation(s)
- Gillian M Hunt
- AIDS Virus Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa.
| | | | | | | | | | | | | |
Collapse
|
229
|
Mild M, Hedskog C, Jernberg J, Albert J. Performance of ultra-deep pyrosequencing in analysis of HIV-1 pol gene variation. PLoS One 2011; 6:e22741. [PMID: 21799940 PMCID: PMC3143174 DOI: 10.1371/journal.pone.0022741] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 07/05/2011] [Indexed: 11/19/2022] Open
Abstract
Introduction Ultra-deep pyrosequencing (UDPS) has been used to detect minority variants within HIV-1 populations. Some aspects of the quality and reproducibility of UDPS have been previously evaluated, but comprehensive studies are still needed. Principal Finding In this study the UDPS technology (FLX platform) was evaluated by analyzing a 120 base pair fragment of the HIV-1 pol gene from plasma samples from two patients and artificial mixtures of molecular clones. UDPS was performed using an optimized experimental protocol and an in-house data cleaning strategy. Nine samples and mixtures were analyzed and the average number of reads per sample was 19,404 (range 8,858–26,846). The two patient plasma samples were analyzed twice and quantification of viral variants was found to be highly repeatable for variants representing >0.27% of the virus population, whereas some variants representing 0.11–0.27% were detected in only one of the two UDPS runs. Bland-Altman analysis showed that a repeated measurement would have a 95% likelihood to lie approximately within ±0.5 log10 of the initial estimate. A similar level of agreement was observed for variant frequency estimates in forward vs. reverse sequencing direction, but here the agreement was higher for common variants than for rare variants. UDPS following PCR amplification with alternative primers indicated that some variants may be incorrectly quantified due to primer-related selective amplification. Finally, the in vitro recombination rate during PCR was evaluated using artificial mixtures of clones and was found to be low. The most abundant in vitro recombinant represented 0.25% of all UDPS reads. Conclusion This study demonstrates that this UDPS protocol results in low experimental noise and high repeatability, which is relevant for future research and clinical use of the UDPS technology. The low rate of in vitro recombination suggests that this UDPS system can be used to study genetic variants and mutational linkage.
Collapse
Affiliation(s)
- Mattias Mild
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | |
Collapse
|
230
|
The evolution of Pyrosequencing® for microbiology: From genes to genomes. J Microbiol Methods 2011; 86:1-7. [DOI: 10.1016/j.mimet.2011.04.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 04/04/2011] [Accepted: 04/05/2011] [Indexed: 12/11/2022]
|
231
|
Delobel P, Saliou A, Nicot F, Dubois M, Trancart S, Tangre P, Aboulker JP, Taburet AM, Molina JM, Massip P, Marchou B, Izopet J. Minor HIV-1 variants with the K103N resistance mutation during intermittent efavirenz-containing antiretroviral therapy and virological failure. PLoS One 2011; 6:e21655. [PMID: 21738752 PMCID: PMC3124548 DOI: 10.1371/journal.pone.0021655] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 06/07/2011] [Indexed: 11/19/2022] Open
Abstract
UNLABELLED The impact of minor drug-resistant variants of the type 1 immunodeficiency virus (HIV-1) on the failure of antiretroviral therapy remains unclear. We have evaluated the importance of detecting minor populations of viruses resistant to non-nucleoside reverse-transcriptase inhibitors (NNRTI) during intermittent antiretroviral therapy, a high-risk context for the emergence of drug-resistant HIV-1. We carried out a longitudinal study on plasma samples taken from 21 patients given efavirenz and enrolled in the intermittent arm of the ANRS 106 trial. Allele-specific real-time PCR was used to detect and quantify minor K103N mutants during off-therapy periods. The concordance with ultra-deep pyrosequencing was assessed for 11 patients. The pharmacokinetics of efavirenz was assayed to determine whether its variability could influence the emergence of K103N mutants. Allele-specific real-time PCR detected K103N mutants in 15 of the 19 analyzable patients at the end of an off-therapy period while direct sequencing detected mutants in only 6 patients. The frequency of K103N mutants was <0.1% in 7 patients by allele-specific real-time PCR without further selection, and >0.1% in 8. It was 0.1%-10% in 6 of these 8 patients. The mutated virus populations of 4 of these 6 patients underwent further selection and treatment failed for 2 of them. The K103N mutant frequency was >10% in the remaining 2, treatment failed for one. The copy numbers of K103N variants quantified by allele-specific real-time PCR and ultra-deep pyrosequencing agreed closely (ρ = 0.89 P<0.0001). The half-life of efavirenz was higher (50.5 hours) in the 8 patients in whom K103N emerged (>0.1%) than in the 11 patients in whom it did not (32 hours) (P = 0.04). Thus ultrasensitive methods could prove more useful than direct sequencing for predicting treatment failure in some patients. However the presence of minor NNRTI-resistant viruses need not always result in virological escape. TRIAL REGISTRATION ClinicalTrials.gov NCT00122551.
Collapse
Affiliation(s)
- Pierre Delobel
- Service des Maladies Infectieuses et Tropicales, Hôpital Purpan, Toulouse, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Abstract
Next-generation sequencing technologies produce a large number of noisy reads from the DNA in a sample. Metagenomics and population sequencing aim to recover the genomic sequences of the species in the sample, which could be of high diversity. Methods geared towards single sequence reconstruction are not sensitive enough when applied in this setting. We introduce a generative probabilistic model of read generation from environmental samples and present Genovo, a novel de novo sequence assembler that discovers likely sequence reconstructions under the model. A nonparametric prior accounts for the unknown number of genomes in the sample. Inference is performed by applying a series of hill-climbing steps iteratively until convergence. We compare the performance of Genovo to three other short read assembly programs in a series of synthetic experiments and across nine metagenomic datasets created using the 454 platform, the largest of which has 311k reads. Genovo's reconstructions cover more bases and recover more genes than the other methods, even for low-abundance sequences, and yield a higher assembly score. Supplementary Material is available at www.liebertoinline.com/cmb .
Collapse
Affiliation(s)
- Jonathan Laserson
- Department of Computer Science, Stanford University, Stanford, California, USA
| | | | | |
Collapse
|
233
|
Isakov O, Modai S, Shomron N. Pathogen detection using short-RNA deep sequencing subtraction and assembly. Bioinformatics 2011; 27:2027-30. [PMID: 21666269 PMCID: PMC3137223 DOI: 10.1093/bioinformatics/btr349] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Early and accurate detection of human pathogen infection is critical for treatment and therapeutics. Here we describe pathogen identification using short RNA subtraction and assembly (SRSA), a detection method that overcomes the requirement of prior knowledge and culturing of pathogens, by using degraded small RNA and deep sequencing technology. We prove our approach's efficiency through identification of a combined viral and bacterial infection in human cells.
Collapse
Affiliation(s)
- Ofer Isakov
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | |
Collapse
|
234
|
Diagnostic Molecular Virology: Current Practice and Future Trends. Mol Microbiol 2011. [DOI: 10.1128/9781555816834.ch34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
235
|
Detection of minority resistance during early HIV-1 infection: natural variation and spurious detection rather than transmission and evolution of multiple viral variants. J Virol 2011; 85:8359-67. [PMID: 21632754 DOI: 10.1128/jvi.02582-10] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Reports of a high frequency of the transmission of minority viral populations with drug-resistant mutations (DRM) are inconsistent with evidence that HIV-1 infections usually arise from mono- or oligoclonal transmission. We performed ultradeep sequencing (UDS) of partial HIV-1 gag, pol, and env genes from 32 recently infected individuals. We then evaluated overall and per-site diversity levels, selective pressure, sequence reproducibility, and presence of DRM and accessory mutations (AM). To differentiate biologically meaningful mutations from those caused by methodological errors, we obtained multinomial confidence intervals (CI) for the proportion of DRM at each site and fitted a binomial mixture model to determine background error rates for each sample. We then examined the association between detected minority DRM and the virologic failure of first-line antiretroviral therapy (ART). Similar to other studies, we observed increased detection of DRM at low frequencies (average, 0.56%; 95% CI, 0.43 to 0.69; expected UDS error, 0.21 ± 0.08% mutations/site). For 8 duplicate runs, there was variability in the proportions of minority DRM. There was no indication of increased diversity or selection at DRM sites compared to other sites and no association between minority DRM and AM. There was no correlation between detected minority DRM and clinical failure of first-line ART. It is unlikely that minority viral variants harboring DRM are transmitted and maintained in the recipient host. The majority of low-frequency DRM detected using UDS are likely errors inherent to UDS methodology or a consequence of error-prone HIV-1 replication.
Collapse
|
236
|
Zhang Y, Sun Y. HMM-FRAME: accurate protein domain classification for metagenomic sequences containing frameshift errors. BMC Bioinformatics 2011; 12:198. [PMID: 21609463 PMCID: PMC3115854 DOI: 10.1186/1471-2105-12-198] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 05/24/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Protein domain classification is an important step in metagenomic annotation. The state-of-the-art method for protein domain classification is profile HMM-based alignment. However, the relatively high rates of insertions and deletions in homopolymer regions of pyrosequencing reads create frameshifts, causing conventional profile HMM alignment tools to generate alignments with marginal scores. This makes error-containing gene fragments unclassifiable with conventional tools. Thus, there is a need for an accurate domain classification tool that can detect and correct sequencing errors. RESULTS We introduce HMM-FRAME, a protein domain classification tool based on an augmented Viterbi algorithm that can incorporate error models from different sequencing platforms. HMM-FRAME corrects sequencing errors and classifies putative gene fragments into domain families. It achieved high error detection sensitivity and specificity in a data set with annotated errors. We applied HMM-FRAME in Targeted Metagenomics and a published metagenomic data set. The results showed that our tool can correct frameshifts in error-containing sequences, generate much longer alignments with significantly smaller E-values, and classify more sequences into their native families. CONCLUSIONS HMM-FRAME provides a complementary protein domain classification tool to conventional profile HMM-based methods for data sets containing frameshifts. Its current implementation is best used for small-scale metagenomic data sets. The source code of HMM-FRAME can be downloaded at http://www.cse.msu.edu/~zhangy72/hmmframe/ and at https://sourceforge.net/projects/hmm-frame/.
Collapse
Affiliation(s)
- Yuan Zhang
- Computer Science and Engineering Department, Michigan State University, East Lansing, MI, USA
| | - Yanni Sun
- Computer Science and Engineering Department, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
237
|
Abstract
Throughout the living world, genetic recombination and nucleotide substitution are the primary processes that create the genetic variation upon which natural selection acts. Just as analyses of substitution patterns can reveal a great deal about evolution, so too can analyses of recombination. Evidence of genetic recombination within the genomes of apparently asexual species can equate with evidence of cryptic sexuality. In sexually reproducing species, nonrandom patterns of sequence exchange can provide direct evidence of population subdivisions that prevent certain individuals from mating. Although an interesting topic in its own right, an important reason for analysing recombination is to account for its potentially disruptive influences on various phylogenetic-based molecular evolution analyses. Specifically, the evolutionary histories of recombinant sequences cannot be accurately described by standard bifurcating phylogenetic trees. Taking recombination into account can therefore be pivotal to the success of selection, molecular clock and various other analyses that require adequate modelling of shared ancestry and draw increased power from accurately inferred phylogenetic trees. Here, we review various computational approaches to studying recombination and provide guidelines both on how to gain insights into this important evolutionary process and on how it can be properly accounted for during molecular evolution studies.
Collapse
Affiliation(s)
- Darren P Martin
- Computational Biology Group, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | | |
Collapse
|
238
|
Abstract
Evaluation of: Willerth SM, Pedro HA, Pachter L, Humeau LM, Arkin AP, Schaffer DV: Development of a low bias method for characterizing viral populations using next generation sequencing technology. PLoS ONE 5(10), E13564 (2010). The recent commercialization of high-throughput sequencing technologies has brought about the possibility of characterizing complex viral populations at previously unattainable scales. An important consideration in designing experiments with these technologies is the need for large quantities of input DNA. To accommodate low-input samples, an amplification step is required, usually a PCR, which comes at the risk of introducing a bias that potentially distorts the view of the viral population composition. By implementing a new sample preprocessing protocol, Willerth et al. address these experimental limitations in the context of complete HIV genome sequencing. Moreover, to facilitate the assembly of short and potentially divergent HIV-derived reads, the authors construct a full-length data-specific consensus sequence using a novel bioinformatics approach that capitalizes on the available HIV complete genomes.
Collapse
Affiliation(s)
| | - Philippe Lemey
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| |
Collapse
|
239
|
Baum PD, Young JJ, Zhang Q, Kasakow Z, McCune JM. Design, construction, and validation of a modular library of sequence diversity standards for polymerase chain reaction. Anal Biochem 2011; 411:106-15. [PMID: 21111699 PMCID: PMC3073996 DOI: 10.1016/j.ab.2010.11.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 11/13/2010] [Accepted: 11/17/2010] [Indexed: 12/29/2022]
Abstract
Methods to measure the sequence diversity of polymerase chain reaction (PCR)-amplified DNA lack standards for use as assay calibrators and controls. Here we present a general and economical method for developing customizable DNA standards of known sequence diversity. Standards ranging from 1 to 25,000 sequences were generated by directional ligation of oligonucleotide "words" of standard length and GC content and then amplified by PCR. The sequence accuracy and diversity of the library were validated using AmpliCot analysis (DNA hybridization kinetics) and Illumina sequencing. The library has the following features: (i) pools containing tens of thousands of sequences can be generated from the ligation of relatively few commercially synthesized short oligonucleotides; (ii) each sequence differs from all others in the library at a minimum of three nucleotide positions, permitting discrimination between different sequences by either sequencing or hybridization; (iii) all sequences have identical length, GC content, and melting temperature; (iv) the identity of each standard can be verified by restriction digestion; and (v) once made, the ends of the library may be cleaved and replaced with sequences to match any PCR primer pair. These standards should greatly improve the accuracy and reproducibility of sequence diversity measurements.
Collapse
Affiliation(s)
- Paul D Baum
- University of California, San Francisco, CA 94143, USA.
| | | | | | | | | |
Collapse
|
240
|
Wright CF, Morelli MJ, Thébaud G, Knowles NJ, Herzyk P, Paton DJ, Haydon DT, King DP. Beyond the consensus: dissecting within-host viral population diversity of foot-and-mouth disease virus by using next-generation genome sequencing. J Virol 2011; 85:2266-75. [PMID: 21159860 PMCID: PMC3067773 DOI: 10.1128/jvi.01396-10] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 11/29/2010] [Indexed: 01/03/2023] Open
Abstract
The diverse sequences of viral populations within individual hosts are the starting material for selection and subsequent evolution of RNA viruses such as foot-and-mouth disease virus (FMDV). Using next-generation sequencing (NGS) performed on a Genome Analyzer platform (Illumina), this study compared the viral populations within two bovine epithelial samples (foot lesions) from a single animal with the inoculum used to initiate experimental infection. Genomic sequences were determined in duplicate sequencing runs, and the consensus sequence of the inoculum determined by NGS was identical to that previously determined using the Sanger method. However, NGS revealed the fine polymorphic substructure of the viral population, from nucleotide variants present at just below 50% frequency to those present at fractions of 1%. Some of the higher-frequency polymorphisms identified encoded changes within codons associated with heparan sulfate binding and were present in both foot lesions, revealing intermediate stages in the evolution of a tissue culture-adapted virus replicating within a mammalian host. We identified 2,622, 1,434, and 1,703 polymorphisms in the inoculum and in the two foot lesions, respectively: most of the substitutions occurred in only a small fraction of the population and represented the progeny from recent cellular replication prior to onset of any selective pressures. We estimated the upper limit for the genome-wide mutation rate of the virus within a cell to be 7.8 × 10(-4) per nucleotide. The greater depth of detection achieved by NGS demonstrates that this method is a powerful and valuable tool for the dissection of FMDV populations within hosts.
Collapse
Affiliation(s)
- Caroline F. Wright
- Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey GU24 0NF, United Kingdom, MRC, University of Glasgow Centre for Virus Research, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, Institut National de la Recherche Agronomique (INRA), UMR BGPI, Cirad TA A-54/K, Campus de Baillarguet, 34938 Montpellier Cedex 5, France, The Sir Henry Wellcome Functional Genomics Facility, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Marco J. Morelli
- Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey GU24 0NF, United Kingdom, MRC, University of Glasgow Centre for Virus Research, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, Institut National de la Recherche Agronomique (INRA), UMR BGPI, Cirad TA A-54/K, Campus de Baillarguet, 34938 Montpellier Cedex 5, France, The Sir Henry Wellcome Functional Genomics Facility, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Gaël Thébaud
- Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey GU24 0NF, United Kingdom, MRC, University of Glasgow Centre for Virus Research, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, Institut National de la Recherche Agronomique (INRA), UMR BGPI, Cirad TA A-54/K, Campus de Baillarguet, 34938 Montpellier Cedex 5, France, The Sir Henry Wellcome Functional Genomics Facility, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Nick J. Knowles
- Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey GU24 0NF, United Kingdom, MRC, University of Glasgow Centre for Virus Research, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, Institut National de la Recherche Agronomique (INRA), UMR BGPI, Cirad TA A-54/K, Campus de Baillarguet, 34938 Montpellier Cedex 5, France, The Sir Henry Wellcome Functional Genomics Facility, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Pawel Herzyk
- Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey GU24 0NF, United Kingdom, MRC, University of Glasgow Centre for Virus Research, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, Institut National de la Recherche Agronomique (INRA), UMR BGPI, Cirad TA A-54/K, Campus de Baillarguet, 34938 Montpellier Cedex 5, France, The Sir Henry Wellcome Functional Genomics Facility, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - David J. Paton
- Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey GU24 0NF, United Kingdom, MRC, University of Glasgow Centre for Virus Research, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, Institut National de la Recherche Agronomique (INRA), UMR BGPI, Cirad TA A-54/K, Campus de Baillarguet, 34938 Montpellier Cedex 5, France, The Sir Henry Wellcome Functional Genomics Facility, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Daniel T. Haydon
- Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey GU24 0NF, United Kingdom, MRC, University of Glasgow Centre for Virus Research, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, Institut National de la Recherche Agronomique (INRA), UMR BGPI, Cirad TA A-54/K, Campus de Baillarguet, 34938 Montpellier Cedex 5, France, The Sir Henry Wellcome Functional Genomics Facility, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Donald P. King
- Institute for Animal Health, Ash Road, Pirbright, Woking, Surrey GU24 0NF, United Kingdom, MRC, University of Glasgow Centre for Virus Research, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, Institut National de la Recherche Agronomique (INRA), UMR BGPI, Cirad TA A-54/K, Campus de Baillarguet, 34938 Montpellier Cedex 5, France, The Sir Henry Wellcome Functional Genomics Facility, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
241
|
Selection and characterization of HIV-1 with a novel S68 deletion in reverse transcriptase. Antimicrob Agents Chemother 2011; 55:2054-60. [PMID: 21357304 DOI: 10.1128/aac.01700-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Resistance to human immunodeficiency virus type 1 (HIV-1) represents a significant problem in the design of novel therapeutics and the management of treatment regimens in infected persons. Resistance profiles can be elucidated by defining modifications to the viral genome conferred upon exposure to novel nucleoside reverse transcriptase (RT) inhibitors (NRTI). In vitro testing of HIV-1LAI-infected primary human lymphocytes treated with β-D-2',3'-dideoxy-2',3'-didehydro-5-fluorocytidine (DFC; Dexelvucitabine; Reverset) produced a novel deletion of AGT at codon 68 (S68Δ) alone and in combination with K65R that differentially affects drug response. Dual-approach clone techniques utilizing TOPO cloning and pyrosequencing confirmed the novel S68Δ in the HIV-1 genome. The S68Δ HIV-1 RT was phenotyped against various antiviral agents in a heteropolymeric DNA polymerase assay and in human lymphocytes. Drug susceptibility results indicate that the S68Δ displayed a 10- to 30-fold increase in resistance to DFC, lamivudine, emtricitabine, tenofovir, abacavir, and amdoxovir and modest resistance to stavudine, β-d-2',3'-oxa-5-fluorocytidine, or 9-(β-D-1,3-dioxolan-4-yl)guanine and remained susceptible to 3'-azido-3'-deoxythymidine, 2',3'-dideoxyinosine (ddI), 1-(β-D-dioxolane)thymine (DOT) and lopinavir. Modeling revealed a central role for S68 in affecting conformation of the β3-β4 finger region and provides a rational for the selective resistance. These data indicate that the novel S68Δ is a previously unrecognized deletion that may represent an important factor in NRTI multidrug resistance treatment strategies.
Collapse
|
242
|
D'Aquila RT, Geretti AM, Horton JH, Rouse E, Kheshti A, Raffanti S, Oie K, Pappa K, Ross LL. Tenofovir (TDF)-selected or abacavir (ABC)-selected low-frequency HIV type 1 subpopulations during failure with persistent viremia as detected by ultradeep pyrosequencing. AIDS Res Hum Retroviruses 2011; 27:201-9. [PMID: 20929395 DOI: 10.1089/aid.2010.0077] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Detection of drug resistance is critical for determining antiretroviral treatment options. Ultradeep pyrosequencing (UDPS; 454 Life Sciences) is capable of detecting virus variant subpopulations with much greater sensitivity than population sequencing, which typically has a detection limit around 20%. UDPS of the HIV-1 reverse transcriptase (RT) (amino acids 56-120) was performed to detect the key mutations K65R and L74V associated with tenofovir and abacavir use. Plasma specimens from subjects with persistent rebound viremia following suppression on tenofovir (n = 8) or abacavir (n = 9)-based therapy were studied. Samples from a subject treated with zidovudine/lamivudine/efavirenz with a similar loss of virologic response served as a control. HIV-1 plasma RNA was ≥3.68 log(10) copies/ml at all time points sequenced. The median number of UDPS sequences analyzed/time point was 33,246. Among the eight tenofovir-treated subjects, three showed high-frequency (>20%) RT K65R at the time of failure, whereas one showed low-frequency (<20%) L74V; no low-frequency K65R was detected in these subjects. Among the nine abacavir-treated subjects, three showed low-frequency K65R; no L74V was detected in these patients. No K65R or L74V was detected in the samples from the control subject. At failure, other RT mutations were detected, including low-frequency NNRTI-resistant species detected at ≥1 time point in nine subjects; the key NNRTI mutation K103N, however, was always observed at >20% frequency. Although UDPS is useful in the detection of low-frequency subpopulations with transmitted resistance in antiviral-naive patients, it may have less utility in treatment-experienced patients with persistent viremia on therapy.
Collapse
Affiliation(s)
| | - Anna Marie Geretti
- Royal Free Hampstead NHS Trust and University College London Medical School, London, United Kingdom
| | | | | | - Asghar Kheshti
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Katrina Oie
- GlaxoSmithKline, Research Triangle Park, North Carolina
| | - Keith Pappa
- GlaxoSmithKline, Research Triangle Park, North Carolina
| | - Lisa L. Ross
- GlaxoSmithKline, Research Triangle Park, North Carolina
| |
Collapse
|
243
|
Saunders NA. Application of nanomaterials to arrays for infectious disease diagnosis. Nanomedicine (Lond) 2011; 6:271-80. [DOI: 10.2217/nnm.10.161] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
As our understanding of infectious agents increases it has become possible to identify the biological components of individual strains with greater accuracy. This ability is aiding clinical microbiologists to control both existing and emerging infectious disease problems. Microarray technologies are playing an increasingly important role in supplying relevant test data. Control of the nanoscale structures of the materials used in arrays, including the supports, reagents and analytes, is now essential. New techniques for the manufacture of nanostructured supports is now allowing rapid advances in the fields of multianalyte testing and high-volume DNA sequencing.
Collapse
Affiliation(s)
- Nicholas A Saunders
- Health Protection Agency, Centre for Infections, 61 Colindale Avenue, London NW9 5HT, UK
| |
Collapse
|
244
|
Wirden M, Soulie C, Valantin MA, Fourati S, Simon A, Lambert-Niclot S, Bonmarchand M, Clavel-Osorio C, Marcelin AG, Katlama C, Calvez V. Historical HIV-RNA resistance test results are more informative than proviral DNA genotyping in cases of suppressed or residual viraemia. J Antimicrob Chemother 2011; 66:709-12. [PMID: 21393164 DOI: 10.1093/jac/dkq544] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Resistance genotyping is often requested due to residual HIV viraemia or for treatment optimization, but may be unsuccessful if plasma RNA levels are too low or undetectable. Analyses of proviral HIV-DNA can provide information about the viral reservoir, because integrated DNA reflects both actively and latently infected cells. OBJECTIVES To determine whether proviral DNA is a potential relevant alternative to HIV-RNA for resistance genotyping in this context. METHODS The resistance mutations harboured by the proviral DNA were compared with the cumulative data for all plasma RNA genotypes previously obtained for the patient concerned. We also investigated whether various parameters, such as CD4 count, level of viraemia or drug pressure, affected the results. RESULTS We collected 134 and 141 DNA genotypes with 443 and 462 corresponding RNA sequences for the reverse transcriptase and protease genes, respectively. The mean rates of concordance between DNA and RNA genotypes were 46.7% for nucleoside reverse transcriptase inhibitors (NRTIs), 26.3% for non-NRTIs and 43.7% for protease inhibitors (PIs). Mixtures were detected for most DNA mutations. The rate of concordant PI mutations was significantly higher for patients taking PIs at the time of DNA genotyping (48% versus 26%; P=0.004). The other factors studied had no impact. CONCLUSIONS In the context of low or undetectable viraemia, it is difficult to reach the archived mutated DNA. Classical RNA genotyping during previous periods of virological failure remains the gold standard for documenting resistance mutations and for the monitoring of future treatments.
Collapse
Affiliation(s)
- Marc Wirden
- Department of Virology, Pitié-Salpêtrière Hospital, 83 bvd de l'Hôpital, 75013 Paris, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Voelkerding KV, Dames S, Durtschi JD. Next generation sequencing for clinical diagnostics-principles and application to targeted resequencing for hypertrophic cardiomyopathy: a paper from the 2009 William Beaumont Hospital Symposium on Molecular Pathology. J Mol Diagn 2011; 12:539-51. [PMID: 20805560 DOI: 10.2353/jmoldx.2010.100043] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
During the past five years, new high-throughput DNA sequencing technologies have emerged; these technologies are collectively referred to as next generation sequencing (NGS). By virtue of sequencing clonally amplified DNA templates or single DNA molecules in a massively parallel fashion in a flow cell, NGS provides both qualitative and quantitative sequence data. This combination of information has made NGS the technology of choice for complex genetic analyses that were previously either technically infeasible or cost prohibitive. As a result, NGS has had a fundamental and broad impact on many facets of biomedical research. In contrast, the dissemination of NGS into the clinical diagnostic realm is in its early stages. Though NGS is powerful and can be envisioned to have multiple applications in clinical diagnostics, the technology is currently complex. Successful adoption of NGS into the clinical laboratory will require expertise in both molecular biology techniques and bioinformatics. The current report presents principles that underlie NGS including sequencing library preparation, sequencing chemistries, and an introduction to NGS data analysis. These concepts are subsequently further illustrated by showing representative results from a case study using NGS for targeted resequencing of genes implicated in hypertrophic cardiomyopathy.
Collapse
|
246
|
Prosperi MCF, Prosperi L, Bruselles A, Abbate I, Rozera G, Vincenti D, Solmone MC, Capobianchi MR, Ulivi G. Combinatorial analysis and algorithms for quasispecies reconstruction using next-generation sequencing. BMC Bioinformatics 2011; 12:5. [PMID: 21208435 PMCID: PMC3022557 DOI: 10.1186/1471-2105-12-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Accepted: 01/05/2011] [Indexed: 11/16/2022] Open
Abstract
Background Next-generation sequencing (NGS) offers a unique opportunity for high-throughput genomics and has potential to replace Sanger sequencing in many fields, including de-novo sequencing, re-sequencing, meta-genomics, and characterisation of infectious pathogens, such as viral quasispecies. Although methodologies and software for whole genome assembly and genome variation analysis have been developed and refined for NGS data, reconstructing a viral quasispecies using NGS data remains a challenge. This application would be useful for analysing intra-host evolutionary pathways in relation to immune responses and antiretroviral therapy exposures. Here we introduce a set of formulae for the combinatorial analysis of a quasispecies, given a NGS re-sequencing experiment and an algorithm for quasispecies reconstruction. We require that sequenced fragments are aligned against a reference genome, and that the reference genome is partitioned into a set of sliding windows (amplicons). The reconstruction algorithm is based on combinations of multinomial distributions and is designed to minimise the reconstruction of false variants, called in-silico recombinants. Results The reconstruction algorithm was applied to error-free simulated data and reconstructed a high percentage of true variants, even at a low genetic diversity, where the chance to obtain in-silico recombinants is high. Results on empirical NGS data from patients infected with hepatitis B virus, confirmed its ability to characterise different viral variants from distinct patients. Conclusions The combinatorial analysis provided a description of the difficulty to reconstruct a quasispecies, given a determined amplicon partition and a measure of population diversity. The reconstruction algorithm showed good performance both considering simulated data and real data, even in presence of sequencing errors.
Collapse
Affiliation(s)
- Mattia C F Prosperi
- Clinic of Infectious Diseases, Catholic University of Sacred Heart, Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Ji H, Li Y, Graham M, Liang BB, Pilon R, Tyson S, Peters G, Tyler S, Merks H, Bertagnolio S, Soto-Ramirez L, Sandstrom P, Brooks J. Next-generation sequencing of dried blood spot specimens: a novel approach to HIV drug-resistance surveillance. Antivir Ther 2011; 16:871-8. [DOI: 10.3851/imp1839] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
248
|
Abstract
Genotypic assays to detect HIV drug resistance are recommended for use in the routine clinical care of HIV-infected persons. Genotypic resistance assays have demonstrated good clinical utility, instructing antiretroviral drug selection and improving therapy outcomes. However, a major limitation of clinically available genotypic assays is the inability to detect low-level drug-resistant variants that exist at low levels within the circulating viral population. Recent data from multiple groups have demonstrated that low-level resistant viral variants are clinically important as they can rapidly grow under drug selection pressure and lead to therapy failure. This article will discuss how ultra-deep sequencing and other new sensitive genotyping technologies can be used to detect low-level drug-resistant HIV variants. It will also address the biological and clinical questions facing the field of HIV genotyping: first, the need to better define the level of sensitivity required to detect drug-resistant variants; second, the effects different resistant variants have on treatment response, and; third, the requirement for genotypic assays to provide information on resistance mutation linkage. Finally, the limitations of the new sensitive genotyping methods will be discussed and how these limitations can lead to discordant results between the different technologies.
Collapse
Affiliation(s)
- Arjet Gega
- Yale University School of Medicine & VA CT Healthcare System 135 College Street, Suite 323 New Haven, CT 06510, USA
| | - Michael J Kozal
- Yale University School of Medicine & VA CT Healthcare System 135 College Street, Suite 323 New Haven, CT 06510, USA
| |
Collapse
|
249
|
Archer J, Rambaut A, Taillon BE, Harrigan PR, Lewis M, Robertson DL. The evolutionary analysis of emerging low frequency HIV-1 CXCR4 using variants through time--an ultra-deep approach. PLoS Comput Biol 2010; 6:e1001022. [PMID: 21187908 PMCID: PMC3002995 DOI: 10.1371/journal.pcbi.1001022] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 11/01/2010] [Indexed: 01/09/2023] Open
Abstract
Large-scale parallel pyrosequencing produces unprecedented quantities of sequence data. However, when generated from viral populations current mapping software is inadequate for dealing with the high levels of variation present, resulting in the potential for biased data loss. In order to apply the 454 Life Sciences' pyrosequencing system to the study of viral populations, we have developed software for the processing of highly variable sequence data. Here we demonstrate our software by analyzing two temporally sampled HIV-1 intra-patient datasets from a clinical study of maraviroc. This drug binds the CCR5 coreceptor, thus preventing HIV-1 infection of the cell. The objective is to determine viral tropism (CCR5 versus CXCR4 usage) and track the evolution of minority CXCR4-using variants that may limit the response to a maraviroc-containing treatment regimen. Five time points (two prior to treatment) were available from each patient. We first quantify the effects of divergence on initial read k-mer mapping and demonstrate the importance of utilizing population-specific template sequences in relation to the analysis of next-generation sequence data. Then, in conjunction with coreceptor prediction algorithms that infer HIV tropism, our software was used to quantify the viral population structure pre- and post-treatment. In both cases, low frequency CXCR4-using variants (2.5-15%) were detected prior to treatment. Following phylogenetic inference, these variants were observed to exist as distinct lineages that were maintained through time. Our analysis, thus confirms the role of pre-existing CXCR4-using virus in the emergence of maraviroc-insensitive HIV. The software will have utility for the study of intra-host viral diversity and evolution of other fast evolving viruses, and is available from http://www.bioinf.manchester.ac.uk/segminator/.
Collapse
Affiliation(s)
- John Archer
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Andrew Rambaut
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Bruce E. Taillon
- 454 Life Sciences, Branford, Connecticut, United States of America
| | - P. Richard Harrigan
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marilyn Lewis
- Pfizer Global Research and Development, Sandwich, United Kingdom
| | - David L. Robertson
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
250
|
Uil TG, Vellinga J, de Vrij J, van den Hengel SK, Rabelink MJWE, Cramer SJ, Eekels JJM, Ariyurek Y, van Galen M, Hoeben RC. Directed adenovirus evolution using engineered mutator viral polymerases. Nucleic Acids Res 2010; 39:e30. [PMID: 21138963 PMCID: PMC3061072 DOI: 10.1093/nar/gkq1258] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Adenoviruses (Ads) are the most frequently used viruses for oncolytic and gene therapy purposes. Most Ad-based vectors have been generated through rational design. Although this led to significant vector improvements, it is often hampered by an insufficient understanding of Ad’s intricate functions and interactions. Here, to evade this issue, we adopted a novel, mutator Ad polymerase-based, ‘accelerated-evolution’ approach that can serve as general method to generate or optimize adenoviral vectors. First, we site specifically substituted Ad polymerase residues located in either the nucleotide binding pocket or the exonuclease domain. This yielded several polymerase mutants that, while fully supportive of viral replication, increased Ad’s intrinsic mutation rate. Mutator activities of these mutants were revealed by performing deep sequencing on pools of replicated viruses. The strongest identified mutators carried replacements of residues implicated in ssDNA binding at the exonuclease active site. Next, we exploited these mutators to generate the genetic diversity required for directed Ad evolution. Using this new forward genetics approach, we isolated viral mutants with improved cytolytic activity. These mutants revealed a common mutation in a splice acceptor site preceding the gene for the adenovirus death protein (ADP). Accordingly, the isolated viruses showed high and untimely expression of ADP, correlating with a severe deregulation of E3 transcript splicing.
Collapse
Affiliation(s)
- Taco G Uil
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|