Science Reviews - Biology, 2024, 3(2), 13-21 Martina Elena Tarozzi
13
Next Generation Sequencing Technologies, Bioinfor-
matics and Artificial Intelligence: A Shared Timeline
Martina Elena Tarozzi, PhD
Indipendent researcher. Florence, Tuscany, Italy
https://doi.org/10.57098/SciRevs.Biology.3.2.2
Received June 10, 2024. Revised June 25, 2024. Accepted June 26, 2024
Abstract: This review provides a comprehensive overview of the fast-paced and intertwined evolution of three
pivotal fields: next-generation sequencing (NGS) technologies, bioinformatics, and artificial intelligence (AI).
The paper begins by tracing the development of sequencing technologies and highlights how advancements in
genetic sequencing have led to an explosion of biological data, necessitating the rise of bioinformatics for data
management and analysis. The review next covers the primary steps and methods used in bioinformatic analysis
and concludes by reporting some of the technical and biological challenges in which AI methods have been
applied.
Introduction
Determining the order of nucleic acids in pol-
ynucleotide molecules and its functional meaning
has been a major biological question since the dis-
covery of the molecular structure of DNA in 1953
(1). Only twenty-four years later in 1977, the first
method for DNA sequencing was published by
Fredrick Sanger and colleagues, who developed the
“chain-termination" or dideoxy technique (2). Im-
provement of this groundbreaking method repre-
sented the first generation of DNA sequencing tech-
nologies, which produced reads nearly one kilobase
(kb) in length. The development of additional tech-
niques such as polymerase chain reaction (PCR) (3)
in 1983 and recombinant DNA technologies pro-
vided the means for generating high quantities of
DNA required by first-generation technologies,
triggering the genomic revolution and ultimately
the first draft of the human genome in 2001(4). A
pivotal turning point was achieved in 2005 with the
advent of Next Generation Sequencing (NGS) tech-
nologies (5), allowing for the massive and parallel
sequencing of whole genomes. In the last decade,
sequencing technologies have expanded to include
methods for RNA sequencing (6,7), giving rise to
the transcriptomic field and to methods for unveil-
ing the structural features and environmental-me-
diated modifications of chromatin and DNA (8), the
epigenomics field, and the single-cell omics technol-
ogies starting in 2009 (9).
Artificial intelligence (AI) was born and rap-
idly improved within this same time frame (Figure
1). Here again, the initial milestones date back to the
early 1950s, with the Turing test and the first use of
the term “Artificial Intelligence” at the Dartmouth
Conference by John McCarthy (10). In the following
three decades, the IT sector released groundbreak-
ing innovations, such as the Internet and World
Wide Web, as well as algorithms that laid the foun-
dation for deep learning, like Convolutional Neural
Networks (CNN), Support Vector Machine (SVM)
and Backpropagation (1114). In less than a decade,
the AI Deep Blue was able to outperform human be-
ings in complex tasks such as playing chess. Starting
in 2001, the conjunction of increasingly powerful
computational resources (storage space and pro-
cessing speed) (15) and the biotechnological devel-
opment that led to NGS, increased the potential for
tandem use of AI and biology. NGS enabled a mas-
sive increase in omics data production, necessitat-
ing the development of computational methods
able to handle such data. The complexity of biolog-
ical processes and data provided opportunities and
challenges that machine learning techniques are
well suited to solve. Consequently, starting from
the early 2000s, computational biology became an
increasingly relevant field.
Martina Elena Tarozzi Science Reviews - Biology, 2024, 3(2), 13-21
14
As molecular biology becomes more data-in-
tensive and AI algorithms better able to handle bio-
logical complexity, the interconnection between
these fields is bound to strengthen. In this review,
we cover the main technological features of NGS
technologies and bioinformatic analysis and pro-
vide an overview of current applications of AI on
sequencing data.
Figure 1: Timeline of improvement milestones in genomics and sequencing technologies (blue arrows), informatics and artificial
intelligence (orange arrows) and computational biology (green arrows).
Overview of illumina technology and se-
quencing assays
NGS refers to modern high-throughput se-
quencing technologies that can be applied to DNA
or RNA. Illumina platforms are the NGS technology
most frequently used in research and clinical set-
tings, and will therefore be the focus in this paper.
The sequencing workflow starts with library
preparation, which varies between different omics.
In genomic workflows, DNA is fragmented either
mechanically, enzymatically, or with transposons in
fragments of appropriate length (typically around
400bp). Next, blunt ends at both ends are repaired:
typically, 5’ ends are phosphorylated and 3’ ends
are repaired with Adenine residues. Subsequently,
adapters are ligated to both ends. Depending on the
experimental design, it is then possible to select ge-
nome regions of interest via enrichment (such as the
exome or a more restricted selection of genes in tar-
get sequencing) or retain the entire genome.
RNA sequencing is a versatile high through-
put sequencing technique introduced in 2008 that
allows for the investigation of gene expression, as
well as alternative splicing , allele-specific expres-
sion , variation in linear nucleotide sequence, novel
transcript expression and gene fusion events . The
appropriate library preparation protocol for RNA
sequencing must be considered based on the
study’s questions in order to reduce bias . There are
four main categories of possible library prepara-
tions: i) total RNAseq: all structural, regulatory,
coding and non-coding RNAs are sequenced, ii)
RNAseq with ribosomal RNA reduction: only
rRNAs useful for phylogenic reconstruction are
kept together with regulatory and coding RNAs, iii)
cDNA capture: only coding RNAs are enriched us-
ing probes targeting exon sequences, iv) polyA se-
lection: only mature mRNAs are isolated through
poly-T probes that bind the 3’ poly-A tail of mRNAs.
The third main omic in terms of sequencing of
nucleic acids is epigenomics. Gene expression can
strongly depend on epigenetic regulation in terms
of changes in the accessibility of certain genomic re-
gions through DNA methylation, histone modifica-
tions, three-dimensional chromatin organization,
and post-transcriptional regulation mechanisms.
Each type of epigenetic modification can be meas-
Science Reviews - Biology, 2024, 3(2), 13-21 Martina Elena Tarozzi
15
ured via an experimental procedure performed be-
fore the library preparation described above. A
thorough overview of these complex techniques can
be found in Mehrmohamadi et al, 2021[24].
There are four main categories of epigenomic
assays: i) DNA methylation: normally investigated
with bisulfite-conversion-based libraries, where
only unmethylated cytosines are converted to uracil
(23) ii) histone modifications: mostly studied with
chromatin immunoprecipitation assays (ChIP-seq),
where crosslinked DNA is treated with antibodies
targeting the histone modification of interest and
pulled down (24), iii) chromatin accessibility: stud-
ied with assays based on the fact that open chroma-
tin is more accessible to fragmentation agents, like
digestion enzymes (e.g. DNase-seq) or transposase
(e.g. ATAC-seq)(25), iv) 3D organization: mostly in-
vestigated with Chromatin Conformation Capture
(3C) derived assays (such as 4C-seq, Hi-C, ChIA-
Drop), whereby nuclei DNA is crosslinked, chi-
meric DNA molecules made of genomic regions
close to one another are formed, and the proximal
genomic regions in the 3D space of the nucleus are
measured using pairwise frequencies between ge-
nomic loci (26).
Once the library is loaded into the sequencer,
it is pumped onto a flow-cell where each single-
stranded fragment hybridizes to flow-cell adapters
on both ends forming a “bridge” structure that con-
fers the name to this step called “bridge amplifica-
tion” (27). After cluster generation, sequencing by
synthesis with reversible chain terminators and a
fluorophore corresponding to each of the four nu-
cleotides (A, C, T, and G), or bases, takes place. The
fluorophore wavelength together with its intensity
determines the base call. The acquired optical sig-
nals for each lane of the flow cell are converted into
base call format files (bcl file) that represent the out-
put of sequencing and the first raw input for bioin-
formatic analysis (Figure 2).
Figure 2: Schematic representation of a targeted genomic library preparation workflow and Illumina sequencing reaction. Figure
adapted from images courtesy of www.illumina.com (https://www.illumina.com/science/technology/next-generation-sequenc-
ing.html, https://www.illumina.com/documents/products/techspotlights/techspotlight_sequencing.pdf ).
Bioinformatic analysis
Bioinformatic analysis involves several steps
of computationally intensive data transformations,
each requiring specific tools. Substantial research
has been done to develop reliable software capable
of performing such data transformations and to
make the computational analysis of NGS data re-
producible. For example, package and environment
management systems such as CONDA allow the in-
stallation and management of software developed
in different languages for different operating sys-
tems.
Martina Elena Tarozzi Science Reviews - Biology, 2024, 3(2), 13-21
16
3.1. Primary Data Analysis
The primary analysis of Illumina raw data
consists in demultiplexing bcl data into FASTQ files.
This process transforms the binary raw files ob-
tained from the optic signal acquired during se-
quencing into a text file in which the nucleotide se-
quence is matched by a PHRED score defining the
quality of the nucleotide, or base, call made by the
sequencer. During this step, index sequences con-
tained in each fragment are used to group all calls
belonging to the same sample into a single FASTQ
file. Depending on the sequencer, this step is per-
formed directly on-instrument by Real-Time Anal-
ysis software or afterwards as first step of a com-
mand line pipeline (28).
3.2 Secondary Data Analysis
Secondary analysis of genomic data is typi-
cally a standardized workflow used in diagnostics
and research . It generally consists of three main
steps: mapping onto the reference genome, post-
alignment processing and, if useful, variant calling.
Workflow management systems such as Snake-
make and Nextflow address the need to concate-
nate the steps required by bioinformatic pipelines
while providing an efficient usage of computational
power through process parallelization. The sam-
ple’s sequence is expressed in FASTQ files contain-
ing the nucleotide sequence and quality scores de-
rived from millions of short reads. Quality checks
are typically performed first in secondary analysis.
One of the most common tools for this task is
FASTQC . Adapter reads and base calls with low
quality scores are trimmed from the sequence using
specific tools such as Trimmomatic , fastp or cu-
tadapt . Mapping algorithms are then used to iden-
tify a location in the reference genome that matches
the experimental read generated via sequencing.
Software settings allow for varying degrees of toler-
ance towards base mismatches and extra spaces to
allow for the detection of possible variants. One
commonly used tool for mapping NGS sequences to
a reference genome is the Burrows-Wheeler Align-
ment (BWA). The output of mapping algorithms are
stored as sam (sequence alignment map) files,
which contain all the information surrounding the
mapping procedure in the metadata section, along
with data concerning the mapped genomic region
and mapping. The binary counterpart to a sam file
is a bam file, which represents the type of data that
will undergo further post-alignment processing
and variant calling. Post-alignment processing con-
sists of sorting, marking duplicates and indexing
the bam file. The variant calling step aims to iden-
tify single nucleotide variants and small insertions
and deletions, reported in Variant Call Format (VCF)
files.
3.3 Tertiary Data Analysis
Tertiary analysis of genomic data uses the list
of variants reported in the VCF file to biologically
interpret the data. This component of data analysis
is highly adaptable depending on the experimental
question. The first and most common step is variant
annotation, aimed at obtaining functional infor-
mation about the type of nucleotide substitution
and its predicted effect. A more classic approach is
to consider only the variants that affect the primary
structure of the coded protein, like missense or
truncating variants, especially if in-silico variant
predictor tools describe them as likely pathogenic.
Functional analysis on groups of significant genes is
often performed with network approaches or over-
representation methods. In recent years, data sci-
ence and machine learning methods applied to ge-
nomic data have been a resourceful approach for ac-
quiring a more complete understanding of poly-
genic contributions in complex diseases (39) and in
the context of precision medicine (40,41). Some of
these applications will be further discussed section
5.
5. Applications of ai in genomics and transcriptomics
ML and DL methods have been applied to se-
quencing data covering various research scopes and
topics. Here, we describe some of the most relevant
fields of application, focusing first on the use of ML
and DL for technical issues associated with NGS
data processing and analysis, and then with exam-
ples of how these methods are used to explore open
biological questions. This section aims at providing
common, promising or exemplifying applications
of AI methods on NGS data in biology and bioinfor-
matics and should not be considered a complete
overview of all its possible applications.
5.1 Applications on technical problems
Secondary bioinformatic analysis: variant calling
Science Reviews - Biology, 2024, 3(2), 13-21 Martina Elena Tarozzi
17
The accurate discovery of single nucleotide
variants from billions of short reads remains a chal-
lenging step in bioinformatics because library prep-
aration, sequencing and data processing tools are
error-prone procedures. These issues become even
more apparent when the object of study are low-fre-
quency somatic mutations or when the input DNA
is of lower quality. Most variant callers use statisti-
cal methods (such as logistic regression, hidden
Markov models, naïve Bayes) to model error
sources and to distinguish whether differences be-
tween experimental reads and the reference ge-
nome are caused by true genetic variants or errors.
In recent years, deep learning has been applied to
address variant calling on NGS data: a common ap-
proach is to address the problem as one of image
recognition, where a Deep Neural Network ana-
lyzes sequencing data that are transformed as im-
ages of read pileups of true genotype calls to com-
pute the genotype likelihoods at each locus. Two of
the first and most popular tools of this kind are
DeepVariant (42) and DeNovoCNN (43), with the
latter specifically used to address the identification
of de novo mutations. Both tools showed higher ac-
curacy compared to classical methods. An alterna-
tive approach is presented in HELLO (44), whereby
comparable performances are obtained by design-
ing Deep Neural Networks that examine aligned
reads to predict the status (ref or alt) of each candi-
date allele given the support for that allele in rela-
tion to the support for the remaining alleles at the
genomic site.
Tertiary bioinformatic analysis: Variant effect prediction
Variant Effect Prediction (VEP) are computa-
tional tools that provide a prediction about the func-
tional significance of a single nucleotide variant
(SNV). The growing use of NGS technologies for
advanced diagnostics has increased the need to bet-
ter classify variants of uncertain significance. VEPs
rely on different types of prior knowledge, such as
protein sequence and structural information, evolu-
tionary sequence conservation, functional experi-
ments, epigenomic data and association studies to
produce an effect score for the variant. In super-
vised VEPs, the algorithm is trained on a set of la-
belled SNVs known as benign or dam2aging ac-
cording to previous knowledge to perform a classi-
fication task. Using this prior knowledge, these
methods compute a score expressing the predicted
effect of the variant. Examples of well-performing
supervised VEPs on human samples are SNP&GO
(45), PolyPhen2 (46) and DEOGEN2 (47). Unsuper-
vised methods do not use any labelled data and
usually rely exclusively on the evolutionary conser-
vation of the genomic locus. This group also in-
cludes deep learning methods, like DeepSequence
(48), considered by a recent benchmark study as the
top-performing tool among 46 tested in deep muta-
tion scanning data (49). An example of a semi-su-
pervised deep learning method is the Illumina Pri-
mateAI (50), which has performed well in the study
of rare diseases.
Visualization of high dimensional datasets
NGS data are highly dimensional because
each sample is sequenced simultaneously. The huge
amount of information contained in these data can
represent an obstacle to the identification of its most
meaningful features. Dimensionality reduction
techniques such as PCA, t-SNE and UMAP are used
to identify latent components in the data that are
not easily accessible due to the high number of var-
iables. Data are thus transformed into a lower di-
mensionality while maintaining the relationships
between data points (e.g., samples) as much as pos-
sible. These methods are extremely versatile. For ex-
ample, they can be used in the pre-processing of
bulk RNA-seq data to identify possible outliers and
relevant covariates(51), to search for recurrent pat-
terns on targeted DNA sequencing data in different
classes of samples (52), or to visualize single-cell
RNA sequencing data. In this context, dimensional-
ity reduction techniques coupled with clustering al-
gorithms are used for cell-type identification tasks,
identifying groups of cells that share similar expres-
sion profiles. Another application of these methods
is lineage trajectory inference(53), which involves
the reconstruction of the position of each individual
cell on the lineage trajectory based on scRNA-seq
profiles with different time points, allowing for the
study of dynamic processes such as the cell cycle,
cell differentiation and cell activation.
6. Future Perspectives
In this review, we summarized the crucial as-
pects and timeline of NGS technologies, bioinfor-
matics and AI, highlighted how they are connected
in a holistic process, and explained the potential
revolutionary insights that can be gained from their
Martina Elena Tarozzi Science Reviews - Biology, 2024, 3(2), 13-21
18
concurrent use. Computational and molecular biol-
ogy have and are continuing to advance at an im-
pressive pace. Machine and deep learning, while
relatively recent breakthroughs in the biological
and biomedical fields, will almost certainly play an
increasing role. Substantial investments are being
made by leading technology companies that, to-
gether with academic researchers, are implement-
ing innovative methodologies, software and archi-
tectures tailored specifically to answer biological
questions. Concurrently, we are witnessing the im-
provement of molecular techniques while sequenc-
ing experiments are evolving and becoming more
affordable, as evident by the growing interest in
long-read sequencing. Taken together, these inno-
vations are a new frontier of research and have the
potential to strongly affect our ability understand
and interact with genetic information.
References
1. Watson JD, Crick FHC. Molecular Structure of Nucleic Acids: A Structure for Deoxyribose Nucleic
Acid. Nat 1953 1714356 [Internet]. 1953 Apr 25 [cited 2021 Jul 6];171(4356):7378. Available from:
https://www.nature.com/articles/171737a0
2. Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating inhibitors. Proc Natl
Acad Sci U S A [Internet]. 1977 [cited 2021 Jul 6];74(12):5463. Available from:
/pmc/articles/PMC431765/?report=abstract
3. Mullis K, Faloona F, Scharf S, Saiki R, Horn G, Erlich H. Specific enzymatic amplification of DNA
in vitro: The polymerase chain reaction. Cold Spring Harb Symp Quant Biol. 1986;51(1):26373.
4. JC V, MD A, EW M, PW L, RJ M, GG S, et al. The sequence of the human genome. Science
[Internet]. 2001 Feb 16 [cited 2021 Jul 6];291(5507):130451. Available from:
https://pubmed.ncbi.nlm.nih.gov/11181995/
5. Heather J, Chain B, Heather JM, Chain B. The Sequence of Sequencers: The History of Sequencing
DNA Genomics The sequence of sequencers: The history of sequencing DNA. Genomics [Internet].
2015;107(1):18. Available from: http://dx.doi.org/10.1016/j.ygeno.2015.11.003
6. Nagalakshmi U, Wang Z, Waern K, Shou C, Raha D, Gerstein M, et al. The Transcriptional
Landscape of the Yeast Genome Defined by RNA Sequencing. Science [Internet]. 2008 Jun 6 [cited
2021 Jul 19];320(5881):1344. Available from: /pmc/articles/PMC2951732/
7. SJ E, WB B, L L, PS S. Gene discovery and annotation using LCM-454 transcriptome sequencing.
Genome Res [Internet]. 2007 Jan [cited 2021 Jul 6];17(1):6973. Available from:
https://pubmed.ncbi.nlm.nih.gov/17095711/
8. JFriedman N, Rando OJ. Epigenomics and the structure of the living genome. Genome Res.
2015;25(10):148290.
9. Wang D, Bodovitz S. Single cell analysis: the new frontier in ‘Omics.’ Trends Biotechnol [Internet].
2010 Jun [cited 2021 Jul 6];28(6):281. Available from: /pmc/articles/PMC2876223/
10. McCarthy J, Minsky ML, Rochester N, Shannon CE. A proposal for the Dartmouth summer
research project on artificial intelligence [Internet]. Vol. 27, AI Magazine. 2006 [cited 2022 Oct 12]. p.
124. Available from: http://www-formal.stanford.edu/jmc/history/dartmouth/dartmouth.html
11. LeCun Y, Boser B, Denker JS, Henderson D, Howard RE, Hubbard W, et al. Backpropagation
applied to digit recognition [Internet]. Vol. 1, Neural computation. 1989. p. 54151. Available from:
https://www.ics.uci.edu/~welling/teaching/273ASpring09/lecun-89e.pdf
12. Lecun Y, Bottou E, Bengio Y, Haffner P. Gradient-Based Learning Applied to Document
Recognition. 1998.
13. Rosenblatt F. The perceptron: A probabilistic model for information storage and organization in
the brain. Psychol Rev. 1958;65(6):386408.
Science Reviews - Biology, 2024, 3(2), 13-21 Martina Elena Tarozzi
19
14. Boser BE, Guyon IM, Vapnik VN. Training algorithm for optimal margin classifiers. Proc Fifth
Annu ACM Work Comput Learn Theory. 1992;14452.
15. Muir P, Li S, Lou S, Wang D, Spakowicz DJ, Salichos L, et al. The real cost of sequencing: Scaling
computation to keep pace with data generation. Genome Biol [Internet]. 2016 Mar 23 [cited 2023 Apr
28];17(1):19. Available from: https://genomebiology.biomedcentral.com/articles/10.1186/s13059-
016-0917-0
16. Lister R, O’Malley RC, Tonti-Filippini J, Gregory BD, Berry CC, Millar AH, et al. Highly
Integrated Single-Base Resolution Maps of the Epigenome in Arabidopsis. Cell [Internet]. 2008 May
2 [cited 2021 Jul 19];133(3):523. Available from: /pmc/articles/PMC2723732/
17. U N, Z W, K W, C S, D R, M G, et al. The transcriptional landscape of the yeast genome defined
by RNA sequencing. Science [Internet]. 2008 Jun 6 [cited 2021 Jul 6];320(5881):13449. Available from:
https://pubmed.ncbi.nlm.nih.gov/18451266/
18. Mehmood A, Laiho A, Venäläinen MS, McGlinchey AJ, Wang N, Elo LL. Systematic evaluation
of differential splicing tools for RNA-seq studies. Brief Bioinform [Internet]. 2020 Dec 1 [cited 2021
Jul 20];21(6):205265. Available from: https://academic.oup.com/bib/article/21/6/2052/5648232
19. Castel SE, Levy-Moonshine A, Mohammadi P, Banks E, Lappalainen T. Tools and best practices
for data processing in allelic expression analysis. Genome Biol [Internet]. 2015;16(1):113. Available
from: http://dx.doi.org/10.1186/s13059-015-0762-6
20. Hutchins AP, Poulain S, Fujii H, Miranda-Saavedra D. Discovery and characterization of new
transcripts from RNA-seq data in mouse CD4+ T cells. Genomics. 2012 Nov 1;100(5):30313.
21. Haas BJ, Dobin A, Stransky N, Li B, Yang X, Tickle T, et al. STAR-Fusion: Fast and Accurate Fusion
Transcript Detection from RNA-Seq. bioRxiv [Internet]. 2017 Mar 24 [cited 2021 Jul 20];120295.
Available from: https://www.biorxiv.org/content/10.1101/120295v1
22. Van Dijk EL, Jaszczyszyn Y, Thermes C. Library preparation methods for next-generation
sequencing: Tone down the bias. Exp Cell Res. 2014 Mar 10;322(1):1220.
23. Mehrmohamadi M, Sepehri MH, Nazer N, Norouzi MR. A Comparative Overview of Epigenomic
Profiling Methods. Front Cell Dev Biol. 2021;9(July):114.
24. Landt SG, Marinov GK, Kundaje A, Kheradpour P, Pauli F, Batzoglou S, et al. ChIP-seq guidelines
and practices of the ENCODE and modENCODE consortia. Genome Res [Internet]. 2012 Sep 1 [cited
2022 Oct 27];22(9):181331. Available from: https://genome.cshlp.org/content/22/9/1813.full
25. Buenrostro JD, Wu B, Chang HY, Greenleaf WJ. ATAC-seq: A Method for Assaying Chromatin
Accessibility Genome-Wide. Curr Protoc Mol Biol [Internet]. 2015 [cited 2022 Oct 27];109:21.29.1.
Available from: /pmc/articles/PMC4374986/
26. Lieberman-Aiden E, Van Berkum NL, Williams L, Imakaev M, Ragoczy T, Telling A, et al.
Comprehensive mapping of long-range interactions reveals folding principles of the human
genome. Science (80- ) [Internet]. 2009 Oct 9 [cited 2021 Jul 5];326(5950):28993. Available from:
/pmc/articles/PMC2858594
27. Bentley DR, Balasubramanian S, Swerdlow HP, Smith GP, Milton J, Brown CG, et al. Accurate
Whole Human Genome Sequencing using Reversible Terminator Chemistry. Nature [Internet]. 2008
Nov 11 [cited 2022 Oct 18];456(7218):53. Available from: /pmc/articles/PMC2581791/
28. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the
interpretation of sequence variants: A joint consensus recommendation of the American College of
Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med
[Internet]. 2015 May 8 [cited 2021 Mar 31]; 17(5):40524. Available from:
/pmc/articles/PMC4544753/
Martina Elena Tarozzi Science Reviews - Biology, 2024, 3(2), 13-21
20
29. Alonso CM, Llop M, Sargas C, Pedrola L, Panadero J, Hervás D, et al. Clinical Utility of a Next-
Generation Sequencing Panel for Acute Myeloid Leukemia Diagnostics. J Mol Diagn [Internet]. 2019
Mar 1 [cited 2022 Oct 13];21(2):22840. Available from: https://pubmed.ncbi.nlm.nih.gov/30576870/
30. Di Resta C, Galbiati S, Carrera P, Ferrari M. Next-generation sequencing approach for the
diagnosis of human diseases: open challenges and new opportunities. EJIFCC [Internet]. 2018 Apr 1
[cited 2022 Oct 13];29(1):4. Available from: /pmc/articles/PMC5949614/
31. Bartoletti-Stella A, Tarozzi M, Mengozzi G, Asirelli F, Brancaleoni L, Mometto N, et al. Dementia-
related genetic variants in an Italian population of early-onset Alzheimer’s disease. Front Aging
Neurosci. 2022;14(September):113.
32. Köster J, Rahmann S. Snakemake-a scalable bioinformatics workflow engine. Bioinformatics.
2012.
33. DI Tommaso P, Chatzou M, Floden EW, Barja PP, Palumbo E, Notredame C. Nextflow enables
reproducible computational workflows. Nat Biotechnol 2017 354 [Internet]. 2017 Apr 11 [cited 2023
Mar 2];35(4):3169. Available from: https://www.nature.com/articles/nbt.3820
34. Andrews, Simon, Krueger, Felix , Segonds-Pichon, Anne , Biggins, Laura , Krueger, Christel ,
Wingett S. FastQC [Internet]. Babraham, UK. 2010. Available from:
http://www.bioinformatics.babraham.ac.uk/projects/fastqc
35. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data.
Bioinformatics [Internet]. 2014 Aug 1 [cited 2021 Nov 17];30(15):211420. Available from:
https://academic.oup.com/bioinformatics/article/30/15/2114/2390096 metanalysis of machine
and deep learning-based CRISPR gRNA design tools. RNA Biol. 2020; 17:13
36. Chen S, Zhou Y, Chen Y, Gu J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics
[Internet]. 2018 Sep 1 [cited 2023 Jan 10];34(17):i88490. Available from:
https://academic.oup.com/bioinformatics/article/34/17/i884/5093234
37. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMB-
net.journal [Internet]. 2011 May 2 [cited 2023 Jan 10];17(1):102. Available from: https://journal.emb-
net.org/index.php/embnetjournal/article/view/200/479
38. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioin-
formatics. 2009;
39. Ho DSW, Schierding W, Wake M, Saffery R, O’Sullivan J. Machine learning SNP based prediction
for precision medicine. Front Genet. 2019;10(MAR):267.
40. Dias R, Torkamani A. Artificial intelligence in clinical and genomic diagnostics. Genome Med.
2019;11(1):112.
41. Álvarez-Machancoses Ó, Galiana EJD, Cernea A, de la Viña JF, Fernández-Martínez JL. On the
role of artificial intelligence in genomics to enhance precision medicine. Pharmgenomics Pers Med.
2020;13:10519.
42. Poplin R, Chang PC, Alexander D, Schwartz S, Colthurst T, Ku A, et al. A universal snp and small-
indel variant caller using deep neural networks. Nat Biotechnol. 2018;36(10):983.
43. Khazeeva G, Sablauskas K, van der Sanden B, Steyaert W, Kwint M, Rots D, et al. DeNovoCNN:
a deep learning approach to de novo variant calling in next generation sequencing data. Nucleic Ac-
ids Res. 2022;50(17):e97.
44. Ramachandran A, Lumetta SS, Klee EW, Chen D. HELLO: improved neural network architectures
and methodologies for small variant calling. BMC Bioinformatics [Internet]. 2021;22(1):131. Availa-
ble from: https://doi.org/10.1186/s12859-021-04311-4
Science Reviews - Biology, 2024, 3(2), 13-21 Martina Elena Tarozzi
21
45. Capriotti E, Calabrese R, Fariselli P, Martelli PL, Altman RB, Casadio R. WS-SNPs&GO: a web
server for predicting the deleterious effect of human protein variants using functional annotation.
BMC Genomics [Internet]. 2013;14 Suppl 3(Suppl 3):S6. Available from: http://www.biomedcen-
tral.com/1471-2164/14/S3/S6
46. Adzhubei I, Jordan DM, Sunyaev SR. Predicting Functional Effect of Human Missense Mutations
Using PolyPhen-2. Curr Protoc Hum Genet [Internet]. 2013 [cited 2021
47. Raimondi D, Tanyalcin I, FertCrossed JSD, Gazzo A, Orlando G, Lenaerts T, et al. DEOGEN2:
Prediction and interactive visualization of single amino acid variant deleteriousness in human pro-
teins. Nucleic Acids Res. 2017;45(W1):W2016.
48. Riesselman AJ, Ingraham JB, Marks DS. Deep generative models of genetic variation capture the
effects of mutations. Nat Methods [Internet]. 2018;15(10):81622. Available from:
http://dx.doi.org/10.1038/s41592-018-0138-4
49. Livesey BJ, Marsh JA. Using deep mutational scanning to benchmark variant effect predictors and
identify disease mutations. Mol Syst Biol [Internet]. 2020 Jul 1 [cited 2022 Oct 26];16(7):e9380. Availa-
ble from: https://onlinelibrary.wiley.com/doi/full/10.15252/msb.20199380
50. Sundaram L, Gao H, Padigepati SR, McRae JF, Li Y, Kosmicki JA, et al. Predicting the clinical
impact of human mutation with deep neural networks. Nat Genet 2018 508 [Internet]. 2018 Jul 23
[cited 2022 Oct 27];50(8):116170. Available from: https://www.nature.com/articles/s41588-018-
0167-z
51. Conesa A, Madrigal P, Tarazona S, Gomez-Cabrero D, Cervera A, McPherson A, et al. A survey
of best practices for RNA-seq data analysis. Genome Biol. 2016;17(1):119.
52. Tarozzi M, Bartoletti-Stella A, Dall’Olio D, Matteuzzi T, Baiardi S, Parchi P, et al. Identification of
recurrent genetic patterns from targeted sequencing panels with advanced data science: a case-study
on sporadic and genetic neurodegenerative diseases. BMC Med Genomics 2022 151 [Internet]. 2022
Feb 10 [cited 2022 Feb 25];15(1):112. Available from: https://bmcmedgenomics.biomedcen-
tral.com/articles/10.1186/s12920-022-01173-4
53. Saelens W, Cannoodt R, Todorov H, Saeys Y. A comparison of single-cell trajectory inference
methods. Nat Biotechnol [Internet]. 2019;37(5):54754. Available from:
http://dx.doi.org/10.1038/s41587-019-0071-9