Acessibilidade / Reportar erro

SELAdb: A database of exonic variants in a Brazilian population referred to a quaternary medical center in São Paulo

Abstract

OBJECTIVES:

High-throughput sequencing of genomes, exomes, and disease-focused gene panels is becoming increasingly common for molecular diagnostics. However, identifying a single clinically relevant pathogenic variant among thousands of genetic polymorphisms is a challenging task. Publicly available genomic databases are useful resources to filter out common genetic variants present in the population and enable the identification of each disease-causing variant. Based on our experience applying these technologies at Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil, we recognized that the Brazilian population is not adequately represented in widely available genomic databases.

METHODS:

Here, we took advantage of our 5-year experience as a high-throughput sequencing core facility focused on individuals with putative genetic disorders to build a genomic database that may serve as a more accurate reference for our patient population: SELAdb.

RESULTS/CONCLUSIONS:

Currently, our database comprises a final cohort of 523 unrelated individuals, including patients or family members managed by different clinics of HCFMUSP. We compared SELAdb with other publicly available genomic databases and demonstrated that this population is very heterogeneous, largely resembling Latin American individuals of mixed origin, rather than individuals of pure European ancestry. Interestingly, exclusively through SELAdb, we identified a spectrum of known and potentially novel pathogenic variants in genes associated with highly penetrant Mendelian disorders, illustrating that pathogenic variants circulating in the Brazilian population that is treated in our clinics are underrepresented in other population databases. SELAdb is freely available for public consultation at: http://intranet.fm.usp.br/sela

Next Generation Sequencing; Database; Mendelian Disorders; Brazil; Population Genetics


INTRODUCTION

Advances in high-throughput sequencing technologies in the last decade have enabled systematic genome profiling of thousands of individuals from diverse populations worldwide. Consequently, we now have several publicly available genomic databases that illustrate the profound variability across different ethnic groups, serving as rich resources for investigators seeking to elucidate the molecular basis of human diseases. Indeed, as the cost of next-generation sequencing (NGS) is rapidly declining, NGS-based approaches, including exome sequencing and disease-focused gene panels, are quickly becoming the mainstay of molecular diagnostics.

For example, a clinician who has identified a patient with phenotypic features resembling a particular familial syndrome in the absence of family history may choose to perform exome sequencing using the genomic DNA from this patient, in hopes of identifying a disease-causing genetic variant. However, such an approach routinely results in the identification of thousands of variants. Defining which of these variants is the true disease-causing allele is clinically challenging. An approach that is usually adopted to overcome this obstacle is to filter out common variants in a population. However, even this approach has limited utility, as certain populations are underrepresented in many populational databases, and the clinically relevant allele frequencies remain uncharacterized. At Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil, we adopt NGS-based methods for the diagnosis of putative Mendelian disorders, as a standard practice. However, the most widely utilized databases (e.g. The Genome Aggregation Database (gnomAD) (11. Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536(7616):285-91. https://doi.org/10.1038/nature19057.
https://doi.org/10.1038/nature19057...
), 1000 Genomes (22. Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, et al. A global reference for human genetic variation. Nature. 2015;526(7571):68-74. https://doi.org/10.1038/nature15393.
https://doi.org/10.1038/nature15393...
)) have poor annotation and an underrepresentation of diverse individuals of South American origin (33. Giolo SR, Soler JM, Greenway SC, Almeida MA, de Andrade M, Seidman JG, et al. Brazilian urban population genetic structure reveals a high degree of admixture. Eur J Hum Genet. 2012;20(1):111-6. https://doi.org/10.1038/ejhg.2011.144.
https://doi.org/10.1038/ejhg.2011.144...
).

The population of Brazil, in particular, is comprised of many different ethnic groups (33. Giolo SR, Soler JM, Greenway SC, Almeida MA, de Andrade M, Seidman JG, et al. Brazilian urban population genetic structure reveals a high degree of admixture. Eur J Hum Genet. 2012;20(1):111-6. https://doi.org/10.1038/ejhg.2011.144.
https://doi.org/10.1038/ejhg.2011.144...
,44. Kehdy FS, Gouveia MH, Machado M, Magalhães WC, Horimoto AR, Horta BL, et al. Origin and dynamics of admixture in Brazilians and its effect on the pattern of deleterious mutations. Proc Natl Acad Sci U S A. 2015;112(28):8696-701. https://doi.org/10.1073/pnas.1504447112.
https://doi.org/10.1073/pnas.1504447112...
), and provides a unique opportunity to identify novel uncharacterized disease-causing variants that are unique to this population. Illustrating this point, an unusually high incidence of pediatric adrenocortical tumors in Brazil (nearly twenty times higher than the global incidence) led to the identification of a novel variant in TP53 (p.R337H) present in nearly 0.3% of the Southeastern Brazilian population and accounting for 90% of pediatric adrenal tumors in this area (55. Pinto EM, Billerbeck AE, Villares MC, Domenice S, Mendonça BB, Latronico AC. Founder effect for the highly prevalent R337H mutation of tumor suppressor p53 in Brazilian patients with adrenocortical tumors. Arq Bras Endocrinol Metabol. 2004;48(5):647-50. https://doi.org/10.1590/S0004-27302004000500009.
https://doi.org/10.1590/S0004-2730200400...

6. Palmero EI, Schuler-Faccini L, Caleffi M, Achatz MI, Olivier M, Martel-Planche G, et al. Detection of R337H, a germline TP53 mutation predisposing to multiple cancers, in asymptomatic women participating in a breast cancer screening program in Southern Brazil. Cancer Lett. 2008;261(1):21-5. https://doi.org/10.1016/j.canlet.2007.10.044.
https://doi.org/10.1016/j.canlet.2007.10...
-77. Ribeiro RC, Sandrini F, Figueiredo B, Zambetti GP, Michalkiewicz E, Lafferty AR, et al. An inherited p53 mutation that contributes in a tissue-specific manner to pediatric adrenal cortical carcinoma. Proc Natl Acad Sci U S A. 2001;98(16):9330-5. https://doi.org/10.1073/pnas.161479898.
https://doi.org/10.1073/pnas.161479898...
). Subsequent studies demonstrated that the high prevalence of p.R337H in this population can be attributed to a founder effect (55. Pinto EM, Billerbeck AE, Villares MC, Domenice S, Mendonça BB, Latronico AC. Founder effect for the highly prevalent R337H mutation of tumor suppressor p53 in Brazilian patients with adrenocortical tumors. Arq Bras Endocrinol Metabol. 2004;48(5):647-50. https://doi.org/10.1590/S0004-27302004000500009.
https://doi.org/10.1590/S0004-2730200400...
), and highlights the critical need of regionally focused population genetics studies. Recently, investigators at the University of São Paulo Institute of Biology developed a database of exomes from 609 elderly individuals, Arquivo Brasileiro Online de Mutações (ABraOM), which serves as a powerful resource for clinicians and investigators researching this group of individuals (88. Naslavsky MS, Yamamoto GL, de Almeida TF, Ezquina SAM, Sunaga DY, Pho N, et al. Exomic variants of an elderly cohort of Brazilians in the ABraOM database. Hum Mutat. 2017;38(7):751-63. https://doi.org/10.1002/humu.23220.
https://doi.org/10.1002/humu.23220...
). Our goal at HCFMUSP was to take advantage of our expertise as an NGS-based facility, with half a decade of experience in using exome sequencing as a diagnostic tool, to create a more representative database of our general patient population.

MATERIALS AND METHODS

Subjects. Our database includes 862 individuals associated with different clinical services (Endocrinology, Neurology, Nephrology, Psychiatric, Gastroenterology, and Rheumatology) at HCFMUSP, comprised of patients with putative Mendelian disorders of uncertain or unknown genetic causes, patients with complex disease traits, patients with sporadic tumors, or unaffected family members. We applied a kinship filter (99. Team H. Hail 0.2.13-81ab564db2b4.) to remove related individuals to produce a final cohort of 523 unrelated Brazilian individuals (240 males, 283 females).

Sequencing. Exome sequencing was performed using the Illumina HiSeq 2500 platform in Escola Superior de Agricultura “Luiz de Queiroz” (ESALQ - 2013-2014) or Laboratório de Sequenciamento em Larga Escala da Faculdade de Medicina da Universidade de São Paulo (SELA-FMUSP - 2014-2019). Library preparation and exome capture were performed using the Nextera Rapid Capture Enrichment (Illumina, San Diego, CA) or the SureSelect Target Enrichment System All Exon +/- UTRs V4, V5, and V6 (Agilent Technologies, Santa Clara, CA), according to the manufacturer's instructions.

Bioinformatics analysis. After quality-control using FastQC(1010. Andrews S. FastQC: A quality control tool for high throughput sequence data. Available from: https://www.bioinformatics.babraham.ac.uk/projects/fastqc/
https://www.bioinformatics.babraham.ac.u...
) and adapter sequencing removal using the bbduk tool from bbmap (1111. Bushnell B. BBMap: A Fast, Accurate, Splice-Aware Aligner. Conference: 9th Annual Genomics of Energy & Environment Meeting, Walnut Creek, CA, March 17-20, 2014.), paired-end reads were aligned to the hg19/GRCh37+decoy version of the human genome using bwa-mem (1212. Li H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv. 2013.). Aligned reads were then coordinate-sorted, deduplicated, and indexed using bamsort and bammarkduplicate tools from biobambam2 (1313. Tischler G, Leonard S. biobambam: tools for read pair collation based algorithms on BAM files. Source Code Biol Med. 2014;9:13. https://doi.org/10.1186/1751-0473-9-13.
https://doi.org/10.1186/1751-0473-9-13...
). Sequencing errors, duplication rates, and coverage metrics were assessed using qualimap (1414. Okonechnikov K, Conesa A, Garcia-Alcalde F. Qualimap 2: advanced multi-sample quality control for high-throughput sequencing data. Bioinformatics. 2016;32(2):292-4. https://doi.org/10.1093/bioinformatics/btv566.
https://doi.org/10.1093/bioinformatics/b...
). For variant calling, we used an incremental joint variant calling strategy based on freebayes (1515. Garrison E, Marth G. Haplotype-based variant detection from short-read sequencing. arXiv. 2012.), as follows: first, we used freebayes to perform variant calling on each bam file, individually. We filtered out low-coverage and low-quality variants (<10x and QUAL<10, respectively). We performed multiallelic sites decomposition, multiallelic variants decomposition, and left normalization of InDels using vt decompose, vcfallelicprimitives (from vcflib), and vt normalize tools (1616. Garrison E. A C++ library for parsing and manipulation VCF files. Available from: https://github.com/vcflib/vcflib
https://github.com/vcflib/vcflib...
,1717. Tan A, Abecasis GR, Kang HM. Unified representation of genetic variants. Bioinformatics. 2015;31(13):2202-4. https://doi.org/10.1093/bioinformatics/btv112.
https://doi.org/10.1093/bioinformatics/b...
). After processing all vcf files, we created a joint list of variants using the vcfoverlay (1616. Garrison E. A C++ library for parsing and manipulation VCF files. Available from: https://github.com/vcflib/vcflib
https://github.com/vcflib/vcflib...
) tool from vcflib. We then used this joint list as an input for freebayes (using the -@ option) to perform a second round of variant calling. We used the “-l” switch from freebayes to restrict calls only to positions of variants reported in the joint list. This final step enabled us to report all sites from the joint list, even in the samples bearing the reference allele. Sex-specific ploidy was set to regions outside the pseudo-autosomal region of the Y chromosome. Next, we used bcftools merge (1818. Li H. A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data. Bioinformatics. 2011;27(21):2987-93. https://doi.org/10.1093/bioinformatics/btr509.
https://doi.org/10.1093/bioinformatics/b...
) to combine the variant information from individual vcf files into a single file containing the sex-corrected allele frequencies for all the variants detected in the dataset. We then used hail (99. Team H. Hail 0.2.13-81ab564db2b4.) to remove related individuals from the dataset. Finally, we used SNPEff and SNPSift(1919. Cingolani P, Platts A, Wang le L, Coon M, Nguyen T, Wang L, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin). 2012;6(2):80-92. https://doi.org/10.4161/fly.19695.
https://doi.org/10.4161/fly.19695...
,2020. Cingolani P, Patel VM, Coon M, Nguyen T, Land SJ, Ruden DM, et al. Using Drosophila melanogaster as a Model for Genotoxic Chemical Mutational Studies with a New Program, SnpSift. Front Genet. 2012;3:35. https://doi.org/10.3389/fgene.2012.00035.
https://doi.org/10.3389/fgene.2012.00035...
) to annotate these variants according to the genomic loci, the functional consequences on protein-coding genes, and dbSNP membership. A summary of the bioinformatics workflow employed to construct Sequenciamento em Larga Escala database (SELAdb) is depicted in Figure 1. Finally, we used somalier (2121. Pedersen BS, Bhetariya PJ, Brown J, Kravitz SN, Marth G, Jensen R, et al. Somalier: rapid relatedness estimation for cancer and germline studies using efficient genome sketches. Genome Med. 2020;12(1):62. https://doi.org/10.1186/s13073-020-00761-2.
https://doi.org/10.1186/s13073-020-00761...
) to estimate the ancestry of our population in comparison with the populations included in the 1000 Genomes project, using the individuals in 1000 Genomes as a training set. We represented these ancestry data using principal component analysis (PCA) plots built using ggplot2 (2222. Wickham H. ggplot2: Elegant Graphics for Data Analysis: Springer-Verlag New York; 2016.) and used a neural network classifier (2121. Pedersen BS, Bhetariya PJ, Brown J, Kravitz SN, Marth G, Jensen R, et al. Somalier: rapid relatedness estimation for cancer and germline studies using efficient genome sketches. Genome Med. 2020;12(1):62. https://doi.org/10.1186/s13073-020-00761-2.
https://doi.org/10.1186/s13073-020-00761...
) to calculate posterior probabilities for the assignment of a sample in our cohort to the populations defined in 1000 Genomes.

Figure 1
Flow chart of data processing steps used to generate SELAdb database.

Ethics

This study was approved by the Institutional Review Board of HCFMUSP, in accordance with the provisions in the Helsinki Declaration. Written informed consent was obtained from all individuals included in SELAdb.

RESULTS

SELAdb accurately captures the diversity of the southeastern Brazilian population

As detailed in Figure 1, we analyzed exome sequencing data from 862 individuals to generate a final cohort of exomes from 523 unrelated individuals (240 males, 283 females) and a corresponding list of annotated variants with allele frequencies, which we deposited in a novel Brazilian population database, SELAdb. Among these 523 individuals, we identified 1,788,789 variants, including 1,615,436 single-nucleotide variants, 47,805 small insertions, 121,255 small deletions, and 4,293 complex substitutions (Table 1). Among these variants, 502,738 (28.1%) are not reported in dbSNP151 and, therefore, may represent novel variants present in the Brazilian population (Table 1). A total of 2,973,280 effects (alterations resulting in potential changes in protein structure, function, and/or expression, such as amino acid changes or abrogation of transcription factor binding sites) could be attributed to these 1,788,789 variants, including 581,757 targeting annotated exons. Included among these are 152,984 synonymous, 207,854 missense, 3,742 stop-gained, 256 stop-lost, 405 start-lost, and 9,653 frameshift variants (Table 2).

Table 1
SELAdb variants classified by type and presence in other databases.

Table 2
Number of effects attributed to variants in SELAdb by region and type.

Then, we sought to determine the ancestry of the individuals in our cohort using the populations defined by the 1000 Genomes project as a reference. The biplot shown in Figure 2 represents the first two principal components that capture most of the variability present in the data. The European (EUR), African (AFR), and East Asian (EAS) populations encompass three well-defined clusters in the extremes of this two-dimensional space, forming the vertices of a triangle. As expected, the admixed American population (AMR) exhibits higher variability and is dispersed over a larger area of the plot, with limbs extending to the areas delimited by the EUR, AFR, and EAS populations (Figure 2). Similar to previous studies, our results indicate that SELAdb, comprising urban southeastern Brazilian individuals, forms a continuum between EUR and AFR populations, which is consistent with a high degree of intermarriage between these two populations in southeastern Brazil (33. Giolo SR, Soler JM, Greenway SC, Almeida MA, de Andrade M, Seidman JG, et al. Brazilian urban population genetic structure reveals a high degree of admixture. Eur J Hum Genet. 2012;20(1):111-6. https://doi.org/10.1038/ejhg.2011.144.
https://doi.org/10.1038/ejhg.2011.144...
). Furthermore, we observe here that individuals in SELAdb broadly overlap with a large fraction of the AMR population (Figure 2). To better quantify and define the putative ancestry of individuals in SELAdb, we applied a neural network classifier to fit individuals in SELAdb to pre-existing EAS, SAS, EUR, AFR, and AMR categories in 1000 Genomes (Figure 3). This analysis revealed that the majority of individuals in SELAdb could, indeed, be classified as AMR (75.5%), followed by EUR (18.0%), AFR (5.2%), and EAS (1.3%) (Figure 3B).

Figure 2
Biplot depicting the two principal components (x-axis: PC1, y-axis: PC2) capturing the highest genetic variation among different populations from 1000 Genomes (colored circles) and SELAdb individuals (red crosses). Each circle or cross represents a single individual. The populations represented by the colored circles are African (AFR), Ad-Mixed American (AMR), East Asian (EAS), European (EUR), and South Asian (SAS).
Figure 3
Classification of SELAdb individuals using a neural network classifier and 1000 Genomes populations as the training set. Panel A depicts a series of biplots (x-axis: PC1, y-axis: PC2) representing the PCA analysis of SELAdb individuals, which are color-coded according to the posterior probabilities of being classified as a distinct population by using 1000 Genomes, including AFR (brown, upper left), AMR (blue, upper right), EAS (yellow, lower left), and EUR (green, lower right). Panel B depicts a biplot (left; x-axis: PC1, y-axis: PC2) representing each SELAdb individual, which is color-coded according the final classification given by the neural network classifier. The final distribution of individuals in each category are represented in the donut plot (right). Consistent with the distribution of SELAdb individuals in Panel A, most SELAdb individuals are classified as AMR.

The AMR population in 1000 Genomes is a heterogeneous group comprised of individuals from different geographic regions of Latin America, but does not include any Brazilian individuals (22. Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, et al. A global reference for human genetic variation. Nature. 2015;526(7571):68-74. https://doi.org/10.1038/nature15393.
https://doi.org/10.1038/nature15393...
). Given that the majority of individuals in SELAdb is classified as AMR, despite the absence of Brazilian individuals in this set, we sought to determine which subgroup of the AMR population in 1000 Genomes is the most similar to individuals from SELAdb. To address this question, we performed a similar PCA analysis, but this time with the AMR population separated into its constituent subpopulations, including Peruvians of Lima (PEL), Mexicans of Los Angeles (MXL), Colombians (CLM), and Puerto Ricans (PUR) (Figure 4A). From this analysis, we can appreciate that the AMR population is comprised of unique subclusters, in which PEL, MXL, CLM, and PUR populations cluster separately within the region spanned by AMR in Figure 2. This observation suggests that the broad space encompassed by AMR in the biplot may be partially explained by the number of populations of diverse origins defined as AMR, rather than by a broad heterogeneity dispersed throughout each population. Indeed, Figure 4A illustrates that the MXL and PEL form a distinct cluster that localizes closer to EAS, and farther from EUR and AFR; this observation is consistent with the theory of the Asian origin of native American populations (2323. Reich D, Patterson N, Campbell D, Tandon A, Mazieres S, Ray N, et al. Reconstructing Native American population history. Nature. 2012;488(7411):370-4. https://doi.org/10.1038/nature11258.
https://doi.org/10.1038/nature11258...
). In contrast, PUR exhibit a distinct pattern, characterized by a higher influence of both EUR and AFR. Finally, CLM exhibit characteristics of both MXL/PEL and PUR. We observe here that SELAdb largely overlaps with PUR, which is consistent with the stronger influence of EUR and AFR in the southeastern Brazilian population (33. Giolo SR, Soler JM, Greenway SC, Almeida MA, de Andrade M, Seidman JG, et al. Brazilian urban population genetic structure reveals a high degree of admixture. Eur J Hum Genet. 2012;20(1):111-6. https://doi.org/10.1038/ejhg.2011.144.
https://doi.org/10.1038/ejhg.2011.144...
). By further quantifying this overlap using a neural network classifier and the subdivided AMR groups (Figure 4B), we could observe that the majority of individuals in SELAdb are classified as PUR (60.2%). This observation suggests a similar contribution of European and African ancestries to both PUR and the southeastern Brazilian populations, which is consistent with common historical aspects of colonization of both geographical regions and recent studies defining the ancestral contributions to each population (33. Giolo SR, Soler JM, Greenway SC, Almeida MA, de Andrade M, Seidman JG, et al. Brazilian urban population genetic structure reveals a high degree of admixture. Eur J Hum Genet. 2012;20(1):111-6. https://doi.org/10.1038/ejhg.2011.144.
https://doi.org/10.1038/ejhg.2011.144...
,2424. Szpiech ZA, Mak ACY, White MJ, Hu D, Eng C, Burchard EG, et al. Ancestry-Dependent Enrichment of Deleterious Homozygotes in Runs of Homozygosity. Am J Hum Genet. 2019;105(4):747-62. https://doi.org/10.1016/j.ajhg.2019.08.011.
https://doi.org/10.1016/j.ajhg.2019.08.0...
).

Figure 4
Panel A: biplot (x-axis: PC1, y-axis: PC2) showing the overlap between 1000 Genomes (colored circles) and SELAdb individuals (red crosses). In this representation, the AMR population is split according to its subpopulations, including Colombians (CLM), Mexicans of Los Angeles (MXL), Peruvians of Lima (PEL), and Puerto Ricans (PUR). Panel B: Neural network classification of SELAdb individuals using the 1000 Genomes as a training set. Consistent with the distribution of SELAdb individuals in Panel A, most SELAdb individuals classify as PUR.

SELAdb enables identification of novel, potentially disease-causing variants in a Brazilian population

Given the ability of SELAdb to identify novel variants present in the Brazilian population, that are distinct from other population databases (Table 1), we sought to evaluate its utility in identifying well documented pathogenic variants in a set of 60 genes associated with highly penetrant genetic disorders according to the American College of Medical Genetics and Genomics (ACMG SF v2.0) (2525. Kalia SS, Adelman K, Bale SJ, Chung WK, Eng C, Evans JP, et al. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics. Genet Med. 2017;19(2):249-55. https://doi.org/10.1038/gim.2016.190.
https://doi.org/10.1038/gim.2016.190...
). We were particularly interested in this set of genes, as variants are related with a high risk of diseases associated with early mortality, including cardiovascular disease and familial neoplasia syndromes. Therefore, we suspected that many of these variants may be unique to the Brazilian population but absent from ABraOM database, given its inclusion criteria (88. Naslavsky MS, Yamamoto GL, de Almeida TF, Ezquina SAM, Sunaga DY, Pho N, et al. Exomic variants of an elderly cohort of Brazilians in the ABraOM database. Hum Mutat. 2017;38(7):751-63. https://doi.org/10.1002/humu.23220.
https://doi.org/10.1002/humu.23220...
).

We identified 24 known pathogenic or likely pathogenic variants according to ClinVar (2626. Landrum MJ, Lee JM, Benson M, Brown GR, Chao C, Chitipiralla S, et al. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 2018;46(D1):D1062-D1067. https://doi.org/10.1093/nar/gkx1153
https://doi.org/10.1093/nar/gkx1153...
) (Table 3). Interestingly, 11 of these variants are exclusive to SELAdb, not being reported in gnomAD and ABraOM. Thirteen variants were also reported in gnomAD and/or ABraOM (7 in gnomAD and ABraOM, 6 in gnomAD only, 0 in ABraOM only). In addition, we identified 7 variants that were predicted to be pathogenic, according to InterVar (2727. Li Q, Wang K. InterVar: Clinical Interpretation of Genetic Variants by the 2015 ACMG-AMP Guidelines. Am J Hum Genet. 2017;100(2):267-80. https://doi.org/10.1016/j.ajhg.2017.01.004.
https://doi.org/10.1016/j.ajhg.2017.01.0...
), among which only 4 were present in gnomAD and/or ABraOM (Table 4). These observations highlight the unique contribution of SELAdb in augmenting the spectrum of potential disease-causing variants present in the Brazilian population and illustrate its clinical and research utility.

Table 3
SELAdb variants classified as pathogenic/likely pathogenic using ClinVar and recommended to be reported by ACMG.

Table 4
SELAdb variants that were classified as pathogenic using InterVar and recommended to be reported by ACMG.

DISCUSSION

The Brazilian population is highly admixed, being comprised of several different ethnic groups, and inadequately represented in publicly available genomic databases (33. Giolo SR, Soler JM, Greenway SC, Almeida MA, de Andrade M, Seidman JG, et al. Brazilian urban population genetic structure reveals a high degree of admixture. Eur J Hum Genet. 2012;20(1):111-6. https://doi.org/10.1038/ejhg.2011.144.
https://doi.org/10.1038/ejhg.2011.144...
,44. Kehdy FS, Gouveia MH, Machado M, Magalhães WC, Horimoto AR, Horta BL, et al. Origin and dynamics of admixture in Brazilians and its effect on the pattern of deleterious mutations. Proc Natl Acad Sci U S A. 2015;112(28):8696-701. https://doi.org/10.1073/pnas.1504447112.
https://doi.org/10.1073/pnas.1504447112...
). Here, we took advantage of our 5-year experience as a large-scale sequencing core facility to build a representative local genomic database for the southeastern Brazilian population, SELAdb. Although many individuals included in SELAdb are patients or family members with rare Mendelian disorders, contributing to the identification of novel disease-causing variants (2828. Chamberlin A, Huether R, Machado AZ, Groden M, Liu HM, Upadhyay K, et al. Mutations in MAP3K1 that cause 46,XY disorders of sex development disrupt distinct structural domains in the protein. Hum Mol Genet. 2019;28(10):1620-8. https://doi.org/10.1093/hmg/ddz002.
https://doi.org/10.1093/hmg/ddz002...

29. de Bruin C, Finlayson C, Funari MF, Vasques GA, Lucheze Freire B, Lerario AM, et al. Two Patients with Severe Short Stature due to a FBN1 Mutation (p.Ala1728Val) with a Mild Form of Acromicric Dysplasia. Horm Res Paediatr. 2016;86(5):342-8. https://doi.org/10.1159/000446476.
https://doi.org/10.1159/000446476...

30. Correa FA, Jorge AA, Nakaguma M, Canton AP, Costa SS, Funari MF, et al. Pathogenic copy number variants in patients with congenital hypopituitarism associated with complex phenotypes. Clin Endocrinol. 2018;88(3):425-31. https://doi.org/10.1111/cen.13535.
https://doi.org/10.1111/cen.13535...

31. França MM, Funari MFA, Lerario AM, Nishi MY, Pita CC, Fontenele EGP, et al. A novel homozygous 1-bp deletion in the NOBOX gene in two Brazilian sisters with primary ovarian failure. Endocrine. 2017;58(3):442-7. https://doi.org/10.1007/s12020-017-1459-2.
https://doi.org/10.1007/s12020-017-1459-...

32. França MM, Lerario AM, Funari MFA, Nishi MY, Narcizo AM, de Mello MP, et al. A Novel Homozygous Missense FSHR Variant Associated with Hypergonadotropic Hypogonadism in Two Siblings from a Brazilian Family. Sex Dev. 2017;11(3):137-42. https://doi.org/10.1159/000477193.
https://doi.org/10.1159/000477193...

33. França MM, Funari MFA, Nishi MY, Narcizo AM, Domenice S, Costa EMF, et al. Identification of the first homozygous 1-bp deletion in GDF9 gene leading to primary ovarian insufficiency by using targeted massively parallel sequencing. Clin Genet. 2018;93(2):408-11. https://doi.org/10.1111/cge.13156.
https://doi.org/10.1111/cge.13156...

34. França MM, Nishi MY, Funari MFA, Lerario AM, Baracat EC, Hayashida SAY, et al. Two rare loss-of-function variants in the STAG3 gene leading to primary ovarian insufficiency. Eur J Med Genet. 2019;62(3):186-9. https://doi.org/10.1016/j.ejmg.2018.07.008.
https://doi.org/10.1016/j.ejmg.2018.07.0...

35. Freire BL, Homma TK, Funari MFA, Lerario AM, Leal AM, Velloso EDRP, et al. Homozygous loss of function BRCA1 variant causing a Fanconi-anemia-like phenotype, a clinical report and review of previous patients. Eur J Med Genet. 2018;61(3):130-3. https://doi.org/10.1016/j.ejmg.2017.11.003.
https://doi.org/10.1016/j.ejmg.2017.11.0...

36. Hisado-Oliva A, Ruzafa-Martin A, Sentchordi L, Funari MFA, Bezanilla-López C, Alonso-Bernáldez M, et al. Mutations in C-natriuretic peptide (NPPC): a novel cause of autosomal dominant short stature. Genet Med. 2018;20(1):91-7. https://doi.org/10.1038/gim.2017.66.
https://doi.org/10.1038/gim.2017.66...

37. Lessel D, Gehbauer C, Bramswig NC, Schluth-Bolard C, Venkataramanappa S, van Gassen KLI, et al. BCL11B mutations in patients affected by a neurodevelopmental disorder with reduced type 2 innate lymphoid cells. Brain. 2018;141(8):2299-311. https://doi.org/10.1093/brain/awy173.
https://doi.org/10.1093/brain/awy173...

38. Shinjo SK, Oba-Shinjo SM, Lerario AM, Marie SKN. A Brazilian family with inclusion body myopathy associated with Paget's disease of bone and frontotemporal dementia linked to the VCP pGly97Glu mutation. Clin Rheumatol. 2018;37(4):1129-36. https://doi.org/10.1007/s10067-017-3913-1.
https://doi.org/10.1007/s10067-017-3913-...

39. Vasques GA, Hisado-Oliva A, Funari MF, Lerario AM, Quedas EP, Solberg P, et al. Long-term response to growth hormone therapy in a patient with short stature caused by a novel heterozygous mutation in NPR2. J Pediatr Endocrinol Metab. 2017;30(1):111-6. https://doi.org/10.1515/jpem-2016-0280.
https://doi.org/10.1515/jpem-2016-0280...
-4040. Veloso MP, Neves PDMM, Jorge LB, Dias CB, Yu L, Pinheiro RBB, et al. Female Patient with Alport Syndrome and Concomitant Membranous Nephropathy: Susceptibility or Association of Two Diseases? Nephron. 2017;136(2):158-62. https://doi.org/10.1159/000458710.
https://doi.org/10.1159/000458710...
), our analyses demonstrate that it adequately represents our local patient population. Through ancestry analysis, we observed that the population captured by SELAdb bears diverse genetic influences, which are characteristic of the admixed southeastern Brazilian population, similar to previous reports (33. Giolo SR, Soler JM, Greenway SC, Almeida MA, de Andrade M, Seidman JG, et al. Brazilian urban population genetic structure reveals a high degree of admixture. Eur J Hum Genet. 2012;20(1):111-6. https://doi.org/10.1038/ejhg.2011.144.
https://doi.org/10.1038/ejhg.2011.144...
). In this analysis, we also identified a large overlap between SELAdb individuals and the 1000 Genomes AMR population (Figures 2-3), especially regarding PUR individuals (Figure 4). Taken together, our analyses illustrate the importance of regional population databases in better representing individuals of diverse origin.

Our effort adds value to another recently launched genetic database on Brazilian individuals, ABraOM (88. Naslavsky MS, Yamamoto GL, de Almeida TF, Ezquina SAM, Sunaga DY, Pho N, et al. Exomic variants of an elderly cohort of Brazilians in the ABraOM database. Hum Mutat. 2017;38(7):751-63. https://doi.org/10.1002/humu.23220.
https://doi.org/10.1002/humu.23220...
). However, given the focus of ABraOM on healthy elderly individuals, pathogenic variants that are present in our patient population may be underrepresented. In contrast, our database is focused on a population of patients or family members with putative genetic diseases in whom we have identified a spectrum of known and potentially novel pathogenic variants, as illustrated in Tables 3 and 4. We believe that SELAdb may be informative regarding the prevalence of pathogenic variants in the southeastern Brazilian population and facilitate future genetics studies on Brazilian individuals.

In conclusion, SELAdb is a publicly available database that is representative of our regional patient population. We believe that, in addition to ABraOM, SELAdb will be a valuable resource for investigators using genomics data from the Brazilian population. SELAdb is rapidly increasing in size; updates and improvements, including more detailed phenotypic annotations associated with specific variants, are expected to be implemented every six months. The data can be freely accessed at http://intranet.fm.usp.br/sela.

ACKNOWLEDGMENTS

This work was supported by grants from the Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, Grants 2013/02162-8 and 2014/50137-5).

REFERENCES

  • 1
    Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536(7616):285-91. https://doi.org/10.1038/nature19057
    » https://doi.org/10.1038/nature19057
  • 2
    Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, et al. A global reference for human genetic variation. Nature. 2015;526(7571):68-74. https://doi.org/10.1038/nature15393
    » https://doi.org/10.1038/nature15393
  • 3
    Giolo SR, Soler JM, Greenway SC, Almeida MA, de Andrade M, Seidman JG, et al. Brazilian urban population genetic structure reveals a high degree of admixture. Eur J Hum Genet. 2012;20(1):111-6. https://doi.org/10.1038/ejhg.2011.144
    » https://doi.org/10.1038/ejhg.2011.144
  • 4
    Kehdy FS, Gouveia MH, Machado M, Magalhães WC, Horimoto AR, Horta BL, et al. Origin and dynamics of admixture in Brazilians and its effect on the pattern of deleterious mutations. Proc Natl Acad Sci U S A. 2015;112(28):8696-701. https://doi.org/10.1073/pnas.1504447112
    » https://doi.org/10.1073/pnas.1504447112
  • 5
    Pinto EM, Billerbeck AE, Villares MC, Domenice S, Mendonça BB, Latronico AC. Founder effect for the highly prevalent R337H mutation of tumor suppressor p53 in Brazilian patients with adrenocortical tumors. Arq Bras Endocrinol Metabol. 2004;48(5):647-50. https://doi.org/10.1590/S0004-27302004000500009
    » https://doi.org/10.1590/S0004-27302004000500009
  • 6
    Palmero EI, Schuler-Faccini L, Caleffi M, Achatz MI, Olivier M, Martel-Planche G, et al. Detection of R337H, a germline TP53 mutation predisposing to multiple cancers, in asymptomatic women participating in a breast cancer screening program in Southern Brazil. Cancer Lett. 2008;261(1):21-5. https://doi.org/10.1016/j.canlet.2007.10.044
    » https://doi.org/10.1016/j.canlet.2007.10.044
  • 7
    Ribeiro RC, Sandrini F, Figueiredo B, Zambetti GP, Michalkiewicz E, Lafferty AR, et al. An inherited p53 mutation that contributes in a tissue-specific manner to pediatric adrenal cortical carcinoma. Proc Natl Acad Sci U S A. 2001;98(16):9330-5. https://doi.org/10.1073/pnas.161479898
    » https://doi.org/10.1073/pnas.161479898
  • 8
    Naslavsky MS, Yamamoto GL, de Almeida TF, Ezquina SAM, Sunaga DY, Pho N, et al. Exomic variants of an elderly cohort of Brazilians in the ABraOM database. Hum Mutat. 2017;38(7):751-63. https://doi.org/10.1002/humu.23220
    » https://doi.org/10.1002/humu.23220
  • 9
    Team H. Hail 0.2.13-81ab564db2b4.
  • 10
    Andrews S. FastQC: A quality control tool for high throughput sequence data. Available from: https://www.bioinformatics.babraham.ac.uk/projects/fastqc/
    » https://www.bioinformatics.babraham.ac.uk/projects/fastqc/
  • 11
    Bushnell B. BBMap: A Fast, Accurate, Splice-Aware Aligner. Conference: 9th Annual Genomics of Energy & Environment Meeting, Walnut Creek, CA, March 17-20, 2014.
  • 12
    Li H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv. 2013.
  • 13
    Tischler G, Leonard S. biobambam: tools for read pair collation based algorithms on BAM files. Source Code Biol Med. 2014;9:13. https://doi.org/10.1186/1751-0473-9-13
    » https://doi.org/10.1186/1751-0473-9-13
  • 14
    Okonechnikov K, Conesa A, Garcia-Alcalde F. Qualimap 2: advanced multi-sample quality control for high-throughput sequencing data. Bioinformatics. 2016;32(2):292-4. https://doi.org/10.1093/bioinformatics/btv566
    » https://doi.org/10.1093/bioinformatics/btv566
  • 15
    Garrison E, Marth G. Haplotype-based variant detection from short-read sequencing. arXiv. 2012.
  • 16
    Garrison E. A C++ library for parsing and manipulation VCF files. Available from: https://github.com/vcflib/vcflib
    » https://github.com/vcflib/vcflib
  • 17
    Tan A, Abecasis GR, Kang HM. Unified representation of genetic variants. Bioinformatics. 2015;31(13):2202-4. https://doi.org/10.1093/bioinformatics/btv112
    » https://doi.org/10.1093/bioinformatics/btv112
  • 18
    Li H. A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data. Bioinformatics. 2011;27(21):2987-93. https://doi.org/10.1093/bioinformatics/btr509
    » https://doi.org/10.1093/bioinformatics/btr509
  • 19
    Cingolani P, Platts A, Wang le L, Coon M, Nguyen T, Wang L, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin). 2012;6(2):80-92. https://doi.org/10.4161/fly.19695
    » https://doi.org/10.4161/fly.19695
  • 20
    Cingolani P, Patel VM, Coon M, Nguyen T, Land SJ, Ruden DM, et al. Using Drosophila melanogaster as a Model for Genotoxic Chemical Mutational Studies with a New Program, SnpSift. Front Genet. 2012;3:35. https://doi.org/10.3389/fgene.2012.00035
    » https://doi.org/10.3389/fgene.2012.00035
  • 21
    Pedersen BS, Bhetariya PJ, Brown J, Kravitz SN, Marth G, Jensen R, et al. Somalier: rapid relatedness estimation for cancer and germline studies using efficient genome sketches. Genome Med. 2020;12(1):62. https://doi.org/10.1186/s13073-020-00761-2
    » https://doi.org/10.1186/s13073-020-00761-2
  • 22
    Wickham H. ggplot2: Elegant Graphics for Data Analysis: Springer-Verlag New York; 2016.
  • 23
    Reich D, Patterson N, Campbell D, Tandon A, Mazieres S, Ray N, et al. Reconstructing Native American population history. Nature. 2012;488(7411):370-4. https://doi.org/10.1038/nature11258
    » https://doi.org/10.1038/nature11258
  • 24
    Szpiech ZA, Mak ACY, White MJ, Hu D, Eng C, Burchard EG, et al. Ancestry-Dependent Enrichment of Deleterious Homozygotes in Runs of Homozygosity. Am J Hum Genet. 2019;105(4):747-62. https://doi.org/10.1016/j.ajhg.2019.08.011
    » https://doi.org/10.1016/j.ajhg.2019.08.011
  • 25
    Kalia SS, Adelman K, Bale SJ, Chung WK, Eng C, Evans JP, et al. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics. Genet Med. 2017;19(2):249-55. https://doi.org/10.1038/gim.2016.190
    » https://doi.org/10.1038/gim.2016.190
  • 26
    Landrum MJ, Lee JM, Benson M, Brown GR, Chao C, Chitipiralla S, et al. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 2018;46(D1):D1062-D1067. https://doi.org/10.1093/nar/gkx1153
    » https://doi.org/10.1093/nar/gkx1153
  • 27
    Li Q, Wang K. InterVar: Clinical Interpretation of Genetic Variants by the 2015 ACMG-AMP Guidelines. Am J Hum Genet. 2017;100(2):267-80. https://doi.org/10.1016/j.ajhg.2017.01.004
    » https://doi.org/10.1016/j.ajhg.2017.01.004
  • 28
    Chamberlin A, Huether R, Machado AZ, Groden M, Liu HM, Upadhyay K, et al. Mutations in MAP3K1 that cause 46,XY disorders of sex development disrupt distinct structural domains in the protein. Hum Mol Genet. 2019;28(10):1620-8. https://doi.org/10.1093/hmg/ddz002
    » https://doi.org/10.1093/hmg/ddz002
  • 29
    de Bruin C, Finlayson C, Funari MF, Vasques GA, Lucheze Freire B, Lerario AM, et al. Two Patients with Severe Short Stature due to a FBN1 Mutation (p.Ala1728Val) with a Mild Form of Acromicric Dysplasia. Horm Res Paediatr. 2016;86(5):342-8. https://doi.org/10.1159/000446476
    » https://doi.org/10.1159/000446476
  • 30
    Correa FA, Jorge AA, Nakaguma M, Canton AP, Costa SS, Funari MF, et al. Pathogenic copy number variants in patients with congenital hypopituitarism associated with complex phenotypes. Clin Endocrinol. 2018;88(3):425-31. https://doi.org/10.1111/cen.13535
    » https://doi.org/10.1111/cen.13535
  • 31
    França MM, Funari MFA, Lerario AM, Nishi MY, Pita CC, Fontenele EGP, et al. A novel homozygous 1-bp deletion in the NOBOX gene in two Brazilian sisters with primary ovarian failure. Endocrine. 2017;58(3):442-7. https://doi.org/10.1007/s12020-017-1459-2
    » https://doi.org/10.1007/s12020-017-1459-2
  • 32
    França MM, Lerario AM, Funari MFA, Nishi MY, Narcizo AM, de Mello MP, et al. A Novel Homozygous Missense FSHR Variant Associated with Hypergonadotropic Hypogonadism in Two Siblings from a Brazilian Family. Sex Dev. 2017;11(3):137-42. https://doi.org/10.1159/000477193
    » https://doi.org/10.1159/000477193
  • 33
    França MM, Funari MFA, Nishi MY, Narcizo AM, Domenice S, Costa EMF, et al. Identification of the first homozygous 1-bp deletion in GDF9 gene leading to primary ovarian insufficiency by using targeted massively parallel sequencing. Clin Genet. 2018;93(2):408-11. https://doi.org/10.1111/cge.13156
    » https://doi.org/10.1111/cge.13156
  • 34
    França MM, Nishi MY, Funari MFA, Lerario AM, Baracat EC, Hayashida SAY, et al. Two rare loss-of-function variants in the STAG3 gene leading to primary ovarian insufficiency. Eur J Med Genet. 2019;62(3):186-9. https://doi.org/10.1016/j.ejmg.2018.07.008
    » https://doi.org/10.1016/j.ejmg.2018.07.008
  • 35
    Freire BL, Homma TK, Funari MFA, Lerario AM, Leal AM, Velloso EDRP, et al. Homozygous loss of function BRCA1 variant causing a Fanconi-anemia-like phenotype, a clinical report and review of previous patients. Eur J Med Genet. 2018;61(3):130-3. https://doi.org/10.1016/j.ejmg.2017.11.003
    » https://doi.org/10.1016/j.ejmg.2017.11.003
  • 36
    Hisado-Oliva A, Ruzafa-Martin A, Sentchordi L, Funari MFA, Bezanilla-López C, Alonso-Bernáldez M, et al. Mutations in C-natriuretic peptide (NPPC): a novel cause of autosomal dominant short stature. Genet Med. 2018;20(1):91-7. https://doi.org/10.1038/gim.2017.66
    » https://doi.org/10.1038/gim.2017.66
  • 37
    Lessel D, Gehbauer C, Bramswig NC, Schluth-Bolard C, Venkataramanappa S, van Gassen KLI, et al. BCL11B mutations in patients affected by a neurodevelopmental disorder with reduced type 2 innate lymphoid cells. Brain. 2018;141(8):2299-311. https://doi.org/10.1093/brain/awy173
    » https://doi.org/10.1093/brain/awy173
  • 38
    Shinjo SK, Oba-Shinjo SM, Lerario AM, Marie SKN. A Brazilian family with inclusion body myopathy associated with Paget's disease of bone and frontotemporal dementia linked to the VCP pGly97Glu mutation. Clin Rheumatol. 2018;37(4):1129-36. https://doi.org/10.1007/s10067-017-3913-1
    » https://doi.org/10.1007/s10067-017-3913-1
  • 39
    Vasques GA, Hisado-Oliva A, Funari MF, Lerario AM, Quedas EP, Solberg P, et al. Long-term response to growth hormone therapy in a patient with short stature caused by a novel heterozygous mutation in NPR2. J Pediatr Endocrinol Metab. 2017;30(1):111-6. https://doi.org/10.1515/jpem-2016-0280
    » https://doi.org/10.1515/jpem-2016-0280
  • 40
    Veloso MP, Neves PDMM, Jorge LB, Dias CB, Yu L, Pinheiro RBB, et al. Female Patient with Alport Syndrome and Concomitant Membranous Nephropathy: Susceptibility or Association of Two Diseases? Nephron. 2017;136(2):158-62. https://doi.org/10.1159/000458710
    » https://doi.org/10.1159/000458710

Publication Dates

  • Publication in this collection
    10 Aug 2020
  • Date of issue
    2020

History

  • Received
    8 Apr 2020
  • Accepted
    30 June 2020
Creative Common - by 4.0
This is an Open Access article distributed under the terms of the Creative Commons License (https://creativecommons.org/licenses/by/4.0/) which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is properly cited.
Faculdade de Medicina / USP Rua Dr Ovídio Pires de Campos, 225 - 6 and., 05403-010 São Paulo SP - Brazil, Tel.: (55 11) 2661-6235 - São Paulo - SP - Brazil
E-mail: clinics@hc.fm.usp.br