Review
Copyright ©The Author(s) 2015.
World J Virology. Aug 12, 2015; 4(3): 265-276
Published online Aug 12, 2015. doi: 10.5501/wjv.v4.i3.265
Table 1 A comparative evaluation of the different virus discovery approaches showing advantages and disadvantages associated with them
Classical approaches(Cell culture and infection based)Nucleic acid sequence-dependent amplification approachesNucleic acid sequence-independent amplification approachesNext-generation sequencers-based metagenomic approaches
Requirement of cell culture systemsYes, required for virus particle enrichmentNot requiredNot requiredNot required
Information about the cytopathic effects of the virusYes, could be achieved through cell changesNo information could be achievedNo information could be achievedNo information could be achieved
Requirement of special equipments for purificationYes, Ultracentrifuge/high speed centrifuges, density gradient is required for preparing pure virusNot necessary, semi pure preparations obtained through low speed centrifuges are suitableNot necessary, semi pure preparations obtained through low speed centrifuges are suitableNot necessary, semi pure preparations obtained through low speed centrifuges are suitable
Information about detailed morphological/structural features of the virusYes, could be achieved through Electron/Atomic Force microscopyNo information on virus morphology/structure could be achieved directlyNo information on virus morphology/structure could be achieved directlyNo information on virus morphology/structure could be achieved directly
Time required for virus identificationLong time is required for identification, ranging from days to weeksComparatively faster, days required if cloning and sequencing is involved. Faster with microarray based approachesComparatively faster, virus could be identified within few daysFastest available approach, identification could be done within days and even some times within hours
Requirement of prior knowledge about the virusNot requiredSome information is required regarding genus/family to design primers/probesBeing sequence independent technique, no information is requiredBeing sequence independent technique, no information is required
Dynamic detection rangeVery narrowNarrowWideExtremely wide
Tolerance to non-viral materialsVulnerable to other pathogens capable of infecting cellBeing sequence dependent, less vulnerable to other sequences from host and other pathogensBeing sequence in-dependent, more vulnerable to other sequences from host and other pathogens. Virus enrichment techniques required before analysisBeing sequence in-dependent, more vulnerable to other sequences from host and other pathogens. Virus enrichment techniques required before analysis
Suitability for discovery of new virusesYesLess suitable, good at discovery of genotypes/variants of known virusesYesYes
Suitability during outbreaksNot suitable due to requirement of long timeNot suitable due to requirement of prior sequence informationYes, but still considerable time is required during outbreaksBeing fast, very much suitable in detecting pathogens in an outbreak scenario
Table 2 Important bioinformatics challenges associated with application of next-generation sequencers in viral diagnostics action taken or proposed to overcome challenges
Bioinformatics challenges associated with application of NGS in viral diagnosticsAction taken or proposed to overcome challenges
Generation of huge volumes of data by NGS platforms-“data deluge”Advancement in storage and computation facilities, availability of computer with greater storage and highly powerful processors, cluster/grid computing and cloud computing. Computation facilities needs to be updated with emergence of newer platforms delivering larger datasets
Challenges in uploading data for submission to databases and supercomputing servers for analysisRequirement of uninterrupted and extremely fast networks
Challenges in storage, public archival and ease of accessCreation of specialized data archive such as the Sequence Read Archive by NIH and ENA (European nucleotide Archive) by EBI. Sharing of data within the three major databases (NIH, EBI and DDBJ) for public accessibility
Challenges in analysis and visualization of large volumes of data, beyond the scope of computation facilities available in molecular biology laboratoriesCreation of metagenomic or NGS data analysis pipelines and integrated tool kits, such as those available at NIH-NCBI, EMBL-EBI, MGRAST, CASAVA, MetaVir, Megan, UCSC Genome Browser, BioLinux, etc., availability of cloud computing based servers such as Galaxy
Challenges in alignment, de novo assembly, gene prediction and phylogenetic analyses NGS datasets, especially short read datasetsAvailability of alignment algorithms/programs such as ABySS, ELAND, SOAP, Bowtie, Cloudburst, Zoom, BWA, SHRiMP, MOM, SeqMap, Metagene, Velvet, QSRA, ALLPATHS, EDENA, VCAKE, FragGeneScan, BLAST, GLIMMER, EULER-SR, Avadis, Eagle View, etc.
Interpretation of huge amount of data generated in metagenomic analyses by NGS platformsProper interpretation of analyzed data is of utmost importance to identify newer pathogens as well as their clinical significance