Acessibilidade / Reportar erro

Intervention of next-generation sequencing in diagnosis of Alzheimer’s disease: challenges and future prospects

Intervenção do sequenciamento de nova geração no diagnóstico da doença de Alzheimer: desafios e perspectivas futuras

ABSTRACT

Clinical diagnosis of several neurodegenerative disorders based on clinical phenotype is challenging due to its heterogeneous nature and overlapping disease manifestations. Therefore, the identification of underlying genetic mechanisms is of paramount importance for better diagnosis and therapeutic regimens. With the emergence of next-generation sequencing, it becomes easier to identify all gene variants in the genome simultaneously, with a system-wide and unbiased approach. Presently various bioinformatics databases are maintained on discovered gene variants and phenotypic indications are available online. Since individuals are unique in their genome, evaluation based on their genetic makeup helps evolve the diagnosis, counselling, and treatment process at the personal level. This article aims to briefly summarize the utilization of next-generation sequencing in deciphering the genetic causes of Alzheimer’s disease and address the limitations of whole genome and exome sequencing.

Keywords:
Neurodegenerative Diseases; High-Throughput Nucleotide Sequencing; Alzheimer Disease; Exome Sequencing; Whole Genome Sequencing

RESUMO

O diagnóstico clínico de vários distúrbios neurodegenerativos com base no fenótipo clínico é difícil devido à sua natureza heterogênea e às manifestações da doença que se sobrepõem. Portanto, a identificação dos mecanismos genéticos subjacentes é de suma importância para um melhor diagnóstico e regimes terapêuticos. Com o surgimento do sequenciamento de próxima geração, o diagnóstico se tornou mais acessível com uma abordagem imparcial em todo o sistema para identificar simultaneamente todas as variantes de genes no genoma. Atualmente, vários bancos de dados de bioinformática sobre variantes genéticas descobertas e indicações fenotípicas estão disponíveis online. Uma vez que os indivíduos são únicos em seu genoma, a avaliação com base em sua composição genética ajudou na evolução do processo de diagnóstico, aconselhamento e tratamento em nível pessoal. Este artigo teve como objetivo resumir brevemente a utilização do sequenciamento de próxima geração para decifrar as causas genéticas da doença de Alzheimer (DA) e abordar as limitações do sequenciamento completo do genoma e do exoma.

Palavras-chave:
Doenças Neurodegenerativas; Sequenciamento de Nucleotídeos em Larga Escala; Doença de Alzheimer; Sequenciamento do Exoma; Sequenciamento Completo do Genoma

NEXT-GENERATION SEQUENCING

Sequencing methods, such as Maxam-Gilbert’s chemical degradation method, were first employed to get fragments sequencing, but were soon replaced by the first-generation sequencing techniques such as Sanger’s chain-termination method, due to its hazardous chemical use and toxicity. Sanger sequencing was the popular rapid method used in the late 1900s for the Human Genome Project (HGP) and was also employed to deduce the gene variants responsible for causing disorders11 Heather JM, Chain B. The sequence of sequencers: the history of sequencing DNA. Genomics. 2016;107(1):1-8. https://doi.org/10.1016/j.ygeno.2015.11.003
https://doi.org/10.1016/j.ygeno.2015.11....
,22 van Dijk EL, Jaszczyszyn Y, Naquin D, Thermes C. The third revolution in sequencing technology. Trends Genet. 2018;34(9):666-81. https://doi.org/10.1016/j.tig.2018.05.008
https://doi.org/10.1016/j.tig.2018.05.00...
. These techniques were efficient, but candidate gene selection and sequencing costs were challenging and time-consuming.

By the early 2000s, high-throughput next-generation sequencing (NGS) technologies were developed, making diagnosis easier and hassle-free. NGS is a combination of biology, statistics and information technology that allows massive parallel sequencing of genomes within a relatively short period of time. It achieves tremendous success in microbial genetics, monogenic diseases and complex diseases such as cancer genomics and other multifactorial syndromes. Recently, neurology also adopted the NGS techniques along with other imaging and biochemical methods to gain more expertise in identifying the variants causing disorders33 Shademan B, Avci CB, Nikanfar M, Nourazarian A. Application of next-generation sequencing in neurodegenerative diseases: opportunities and challenges. Neuromolecular Med. 2021;23(2):225-35. https://doi.org/10.1007/s12017-020-08601-7
https://doi.org/10.1007/s12017-020-08601...
. NGS can also be used to study deoxyribonucleic acid (DNA) methylation, protein DNA interaction, ribonucleic acid (RNA) study (RNA-Seq) etc.44 Sun Y, Ruivenkamp CAL, Hoffer MJV, Vrijenhoek T, Kriek M, van Asperen CJ, et al. Next-generation diagnostics: gene panel, exome, or whole genome? Hum Mutat. 2015;36(6):648-55. https://doi.org/10.1002/humu.22783
https://doi.org/10.1002/humu.22783...
.

There are three NGS approaches currently employed namely whole-genome sequencing (WGS), whole-exome sequencing (WES), and gene-targeted panels.

WHOLE GENOME SEQUENCING

WGS sequences the whole genome together. It helps to uncover variation in any part of the human genome, including coding, noncoding, and mitochondrial DNA (mtDNA) regions. WGS is considered the best option once DNA variations outside protein-coding regions can affect gene activity and protein production, potentially leading to genetic disorders44 Sun Y, Ruivenkamp CAL, Hoffer MJV, Vrijenhoek T, Kriek M, van Asperen CJ, et al. Next-generation diagnostics: gene panel, exome, or whole genome? Hum Mutat. 2015;36(6):648-55. https://doi.org/10.1002/humu.22783
https://doi.org/10.1002/humu.22783...
. It also helps to gather more information on an unknown or partially-known disorder and to discover the genomic instabilities leading to complex disorders55 Choudhury A, Ramsay M, Hazelhurst S, Aron S, Bardien S, Botha G, et al. Whole-genome sequencing for an enhanced understanding of genetic variation among South Africans. Nat Commun. 2017;8(1):2062. https://doi.org/10.1038/s41467-017-00663-9
https://doi.org/10.1038/s41467-017-00663...
. It becomes easier to predict any specific variation running in the linage or genetic pool leading to specific phenotypes through various genome-wide association studies (GWAS). In the Encyclopedia of DNA Elements (ENCODE) project, one can see that not only coding regions but also non-coding regions are responsible for causing different complex traits66 ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57-74. https://doi.org/10.1038/nature11247
https://doi.org/10.1038/nature11247...
. WGS allows for the detection of copy number variations (CNVs), gross chromosomal abnormalities, and intergenic, regulatory and deep intronic variants, leading to a higher diagnostic yield. In the neurogenetics field, the WGS was first successfully used for the identification of a causative coding mutation in an autosomal recessive neurodegenerative Charcot-Marie Tooth disease77 Jayadev S, Smith CO, Bird TD. Neurogenetics: five new things. Neurol Clin Pract. 2011;1(1):41-8. https://doi.org/10.1212/CPJ.0b013e31823c0f5f
https://doi.org/10.1212/CPJ.0b013e31823c...
. Whole-genome methylation-specific studies can provide important information on how epigenetic and environmental factors alter gene expression.

WHOLE EXOME SEQUENCING

WES sequences only the exons or protein-coding parts of genes. It is seen that most known disease-causing mutations (∼85%) occur in exons of the gene, hence WES is widely used among clinicians and academics. It is targeted only to exons; therefore, considered a cost-effective method that demands less storage volume (∼4–5 Gb per exome) and reduced time consumption for analysis88 Gao HM, Hong JS. Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression. Trends Immunol. 2008;29(8):357-65. https://doi.org/10.1016/j.it.2008.05.002
https://doi.org/10.1016/j.it.2008.05.002...
. WES offers comprehensive coverage and increased sequencing depth which helps in identifying single nucleotide variants (SNVs) and small insertions/deletions (indels) for population genetics, genetic disease research, and cancer studies. It provides a better platform for detecting mutations running in a family using trio analysis which enables couples to plan their family in a better and healthier way. Through the exome enrichment strategy, we can get a more precise view of gene regulation which includes untranslated regions (UTRs) and microRNAs (miRNA). With WES, there are chances of incidental findings, which can give a valuable insight to the existing knowledge of the disease condition and its pathogenesis in various disorders99 Foo JN, Liu JJ, Tan EK. Whole-genome and whole-exome sequencing in neurological diseases. Nat Rev Neurol. 2012;8(9):508-17. https://doi.org/10.1038/nrneurol.2012.148
https://doi.org/10.1038/nrneurol.2012.14...
. It helps modify disease diagnosis steps and treatment strategies better.

Gene-targeted panels or custom panels sequence only a few genes that are particularly linked to a specific disorder. Gene-targeted panels are observed to be highly effective in the diagnosis of genetic diseases. It is often very small (250 Kb to 5 Mb) in size thus bringing down sequencing requirements and helping in answering distinct scientific questions quickly. It is an economic and suitable application for finding a particular disease or disorder. However, this approach is limited when it comes to complex neurodegenerative disorders.

NEXT-GENERATION SEQUENCING WORKFLOW

There are different techniques and pipelines used in sequencing genomes, depending upon the demands at a specific time. But all the methods notably follow three steps in NGS i.e., library preparation, sequencing, and data analysis. The DNA/RNA is extracted first from the tissue sample, then a quality control (QC) check is done to ensure its purity and quantity by ultraviolet (UV) spectrophotometer and fluorometric methods1010 Singer J, Ruscheweyh HJ, Hofmann AL, Thurnherr T, Singer F, Toussaint NC, et al. NGS-pipe: a flexible, easily extendable and highly configurable framework for NGS analysis. Bioinformatics. 2018;34(1):107-8. https://doi.org/10.1093/bioinformatics/btx540
https://doi.org/10.1093/bioinformatics/b...
.

Template preparation is the prime step in NGS workflow, where the DNA/complementary DNA (cDNA) library is prepared by fragmenting into numerous small coting by physical, enzymatic, and chemical methods, and attaching adaptors to both ends. These libraries are then amplified either by emulsion PCR (ePCR) in ion torrent sequencing or cluster formation by bridge PCR (bPCR) in Illumina sequencing in different customized sizes and prepared for sequencing. The sequenced library can be directly used for whole-genome analysis or undergo a targeted enrichment process for whole-exome analysis and targeted gene panel testing1111 Hess JF, Kohl TA, Kotrová M, Rönsch K, Paprotka T, Mohr V, et al. Library preparation for next generation sequencing: a review of automation strategies. Biotechnol Adv. 2020;41:107537. https://doi.org/10.1016/j.biotechadv.2020.107537
https://doi.org/10.1016/j.biotechadv.202...
.

Most clinical sequencing is performed on different types of instruments such as Illumina sequencers including the HiSeq, MiSeq, NexSeq, Pacific Biosciences, Ion Torrent series of machines including the IonPGM, IonProton, and IonS5, and others1212 Thankachan A, Thomas B. A study of next generation sequencing data, workflow, application and platform comparison. Series: Materials Science and Engineering. International Conference on Recent Advancements and Effectual Researches in Engineering Science and Technology (RAEREST) 20-21 April 2018, Kerala State, India. https://doi.org/10.1088/1757-899X/396/1/012031
https://doi.org/10.1088/1757-899X/396/1/...
. The data generated after sequencing is analyzed using different pipelines and software packages. The results obtained will be interpreted based on the requirement of analysis using various sets of bioinformatics tools.

ALZHEIMER’S DISEASE

Neurodegenerative disorder (NDD), as the name suggests, is a disorder in which cells of the central nervous system stop working or die. They are classified and diagnosed based on clinical features such as physical signs, symptom-onset, and disease course. Alzheimer’s disease (AD) is one of the most common NDDs characterized by dementia that typically begins with subtle mild cognitive impairment (MCI), gradually becomes severe and, finally, leads to total impairment of mental functions. It is commonly seen in the aging population and is becoming a significant cause of socio-economic burden worldwide. Neuropathologic findings mainly extracellular β-amyloid plaques and intraneuronal neurofibrillary tangles (containing tau protein that accumulate in vulnerable brain regions) are the hallmark of AD1313 Lopez JAS, González HM, Léger GC. Alzheimer’s disease. Handb Clin Neurol. 2019;167:231-55. https://doi.org/10.1016/B978-0-12-804766-8.00013-3
https://doi.org/10.1016/B978-0-12-804766...
. Initially, damage occurs in the hippocampus and the entorhinal cortex (memory-forming part of the brain). It then leads to seizure of neuronal function and loose connections of neurons, and gradually to shrinkage of brain parts.

As of 2021, more than 50 million people were affected by dementia worldwide, and this number is estimated to triple to 152 million by 2050 as the world’s population ages1414 Porsteinsson AP, Isaacson RS, Knox S, Sabbagh MN, Rubino I. Diagnosis of early Alzheimer’s disease: clinical practice in 2021. J Prev Alzheimers Dis. 2021;8(3):371-86. https://doi.org/10.14283/jpad.2021.23
https://doi.org/10.14283/jpad.2021.23...
. From 1990 to 2019, the incidence and prevalence of AD and other dementias increased 147.95% and 160.84%, respectively1515 Li X, Feng X, Sun X, Hou N, Han F, Liu Y. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990-2019. Front Aging Neurosci. 2022;14:937486. https://doi.org/10.3389/fnagi.2022.937486
https://doi.org/10.3389/fnagi.2022.93748...
.

Four subtypes are identified in AD so far. Familial or Early-Onset Alzheimer’s Disease (EOAD) constitutes less than 2% of total AD; neurological and depressive behaviors are early symptoms of EOAD1616 Bature F, Guinn BA, Pang D, Pappas Y. Signs and symptoms preceding the diagnosis of Alzheimer’s disease: a systematic scoping review of literature from 1937 to 2016. BMJ Open. 2017;7(8):e015746. https://doi.org/10.1136/bmjopen-2016-015746
https://doi.org/10.1136/bmjopen-2016-015...
. Mutations in amyloid precursor protein (APP), presenin 1 (PSEN1), and presenin 2 (PSEN2), discovered through linkage studies, are the genes predominantly responsible for causing EOAD (Table 1). EOAD is referred to as “Mendelian AD” due to the almost complete penetrance and mostly autosomal-dominant mode of transmission of implicated DNA sequence changes1717 Bertram L. Next generation sequencing in Alzheimer’s disease. Methods Mol Biol. 2016;1303:281-97. https://doi.org/10.1007/978-1-4939-2627-5_17
https://doi.org/10.1007/978-1-4939-2627-...
.

Table 1
Common genes in early onset Alzheimer’s disease.

With the advent of NGS, clinicians can better trace diseases at molecular level. AD and its associated genes have been researched extensively1818 Hippius H, Neundörfer G. The discovery of Alzheimer’s disease. Dialogues Clin Neurosci. 2003;5(1):101-8. https://doi.org/10.31887/DCNS.2003.5.1/hhippius
https://doi.org/10.31887/DCNS.2003.5.1/h...
2222 Weller J, Budson A. Current understanding of Alzheimer’s disease diagnosis and treatment. F1000Res. 2018;7:F1000 Faculty Rev-1161. https://doi.org/10.12688/f1000research.14506.1
https://doi.org/10.12688/f1000research.1...
. In the late 1900s and early 2000s, many genes were found to cause Alzheimer’s phenotype through GWAS. In 2003, the first GWAS were initiated in AD and, in 2007, it was published a meta-analysis of AD susceptibility genes. An AD database was then created, called AlzGene (http://www.alzgene.org)2323 Bertram L, McQueen MB, Mullin K, Blacker D, Tanzi RE. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet. 2007;39(1):17-23. https://doi.org/10.1038/ng1934
https://doi.org/10.1038/ng1934...
.

An estimated 52 pathogenic mutations are identified in the APP gene; most of them are positioned in the vicinity of the β and γ-secretase cleavage sites (exons 16 and 17). Different mutations in V717I/G/F/L and E693K/Q/G/Del residues of APP make them mutation hotspots in the APP gene.

Mutations in exons 5, 6, 7, and 8 of the PSEN1 gene account for 70% of all identified mutations. Five different mutations of PSEN1 residue 143 (I143V/F/N/T/M, encoded by exon 5) are identified, making I143 residue a mutation hotspot2424 Zou Z, Liu C, Che C, Huang H. Clinical genetics of Alzheimer’s disease. Biomed Res Int. 2014;2014:291862. https://doi.org/10.1155/2014/291862
https://doi.org/10.1155/2014/291862...
. It has been discerned that the PSEN1 variant (p.Thr291Pro), found in an individual presenting with spastic paraplegia, can later precede dementia onset in PSEN1-related familial AD2525 Chelban V, Breza M, Szaruga M, Vandrovcova J, Murphy D, Lee CJ, et al. Spastic paraplegia preceding PSEN1-related familial Alzheimer’s disease. Alzheimers Dement (Amst). 2021;13(1):e12186. https://doi.org/10.1002/dad2.12186
https://doi.org/10.1002/dad2.12186...
.

Several candidate gene approaches and GWAS have been performed to identify new genes related to AD (Table 2). Late-onset AD (LOAD) is reported to be caused by multiple gene; semantic dementia and conceptual formation deficit progress in LOAD. Around 90–95% of AD cases are attributed to sporadic mutations2626 Zhang Q, Sidorenko J, Couvy-Duchesne B, Marioni RE, Wright MJ, Goate AM, et.al. Risk prediction of late-onset Alzheimer’s disease implies an oligogenic architecture. Nat Commun. 2020;11(1):4799. https://doi.org/10.1038/s41467-020-18534-1
https://doi.org/10.1038/s41467-020-18534...
. AD overlaps with other disease-related pathways such as Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Huntington disease (HD)2727 Gan L, Cookson MR, Petrucelli L, La Spada AR. Converging pathways in neurodegeneration, from genetics to mechanisms. Nat Neurosci. 2018;21(10):1300-9. https://doi.org/10.1038/s41593-018-0237-7
https://doi.org/10.1038/s41593-018-0237-...
.

Table 2
Common genes in late onset Alzheimer’s disease.

Apolipoprotein E (APOE) e4 allele on chromosome 19, identified using Sanger and family-based approaches, significantly contributes to AD diagnosis in homozygous (APOE e4/e4) and heterozygous (APOE e3/e4) conditions. APOE e4 alleles are strongly associated with AD risk and contribute to various functional abnormalities, neurotoxicity, mitochondrial dysfunction, and cerebrovascular defects2828 Liu CC, Liu CC, Kanekiyo T, Xu H, Bu G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol. 2013;9(2):106-18. https://doi.org/10.1038/nmeurol.2012.263
https://doi.org/10.1038/nmeurol.2012.263...
.

Various studies have been conducted targeting ABCA7, BIN1, CLU, CR1, MS4A6A, EPHA1, CD2AP, and PICALM in different genetic pools. Few pathogenic mutations such as splice site, stop mutation, and frameshift deletions were identified suggesting a loss-of-function mechanism associated with LOAD2929 Bagyinszky E, Youn YC, An SSA, Kim SY. The genetics of Alzheimer’s disease. Clin Interv Aging. 2014;9:535-51. https://doi.org/10.2147/CIA.S51571
https://doi.org/10.2147/CIA.S51571...
. Several missense mutations were found, of which most variants were classified as of uncertain significance due to the lack of functional studies.

A well-known mutation in TREM2 [R47H], identified as causing partial loss of function, contributes to Aβ accumulation by attenuating microglial-mediated Aβ clearance3030 Condello C, Yuan P, Grutzendler J. Microglia-mediated neuroprotection, TREM2, and Alzheimer’s disease: evidence from optical imaging. Biol Psychiatry. 2018;83(4):377-87. https://doi.org/10.1016/j.biopsych.2017.10.007
https://doi.org/10.1016/j.biopsych.2017....
. The clinical phenotype of mutations in FTD genes, including GRID2IP, WDR76, GRN, MAPT, and C9ORF72, can be clinically indistinguishable from typical AD3131 Harms M, Benitez BA, Cairns N, Cooper B, Cooper P, Mayo K, et al. C9orf72 hexanucleotide repeat expansions in clinical Alzheimer disease. JAMA Neurol. 2013;70(6):736-41. https://doi.org/10.1001/2013.jamaneurol.537
https://doi.org/10.1001/2013.jamaneurol....
. Rare variants in the MAPT gene were found to be associated with AD in patients without ApoE e4 and tau pathology3232 Jin SC, Pastor P, Cooper B, Cervantes S, Benitez BA, Razquin C, et al. Pooled-DNA sequencing identifies novel causative variants in PSEN1, GRN and MAPT in a clinical early-onset and familial Alzheimer’s disease Ibero-American cohort. Alzheimers Res Ther. 2012;4(4):34. https://doi.org/10.1186/alzrt137
https://doi.org/10.1186/alzrt137...
. Loss-of-function or null variants in the SORL1 gene is a significant genetic risk factor for AD, as the truncated protein may result in disruption of its ability to bind APP3333 Raghavan NS, Brickman AM, Andrews H, Manly JJ, Schupf N, Lantigua R, et al. Whole-exome sequencing in 20,197 persons for rare variants in Alzheimer’s disease. Ann Clin Transl Neurol. 2018;5(7):832-42. https://doi.org/10.1002/acn3.582
https://doi.org/10.1002/acn3.582...
.

Homozygous and compound heterozygous VWA2 mutations mimic autosomal recessive inheritance in sporadic AD cases3434 Hoogmartens J, Hens E, Engelborghs S, Vandenberghe R, De Deyn PP, Cacace R, et al. Contribution of homozygous and compound heterozygous missense mutations in VWA2 to Alzheimer’s disease. Neurobiol Aging. 2021;99:100.e17-100.e23. https://doi.org/10.1016/j.neurobiolaging.2020.09.009
https://doi.org/10.1016/j.neurobiolaging...
. A missense variant p.Asp238Glu in UNC13B showed segregation within two families of Puerto Rican ancestry and was overrepresented in the AD cases3535 Rajabli F, Feliciano-Astacio BE, Cukier HN, Wang L, Griswold AJ, Hamilton-Nelson KL, et al. Linkage of Alzheimer disease families with Puerto Rican ancestry identifies a chromosome 9 locus. Neurobiol Aging. 2021;104:115.e1-115.e7. https://doi.org/10.1016/j.neurobiolaging.2021.02.019
https://doi.org/10.1016/j.neurobiolaging...
.

A family-based study showed a genome-wide significant linkage peak in 9p21 which overlapped with an AD linkage region. Novel genome-wide significant (GWS) AD-associated non-synonymous variants were identified, as well as a protective variant in PLCG2 (p.P522R), a risk variant in ABI3 (p.S209F), and a novel variant in TREM2 (p.R62H). These genes are highly expressed in microglia and highlight an immune-related protein-protein interaction network enriched for previously identified AD risk genes3636 Sims R, van der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat Genet. 2017;49(9):1373-84. https://doi.org/10.1038/ng.3916
https://doi.org/10.1038/ng.3916...
.

Familial segregation in PLD3 (V232M) was seen, suggesting that PLD3 influences APP metabolism, such that overexpression leads to lower Aβ levels while knock-down of PLD3 leads to increased levels of Aβ3737 Cruchaga C, Karch CM, Jin SC, Benitez BA, Cai Y, Guerreiro R, et al. Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer’s disease. Nature. 2014;505(7484):550-4. https://doi.org/10.1038/nature12825
https://doi.org/10.1038/nature12825...
. The MUC6 VNTR repeat expansion influences AP2A2 gene expression involved in clathrin-coated vesicle function and is associated with AD pathogenesis, particularly tau proteinopathy3838 Nelson PT, Fardo DW, Katsumata Y. The MUC6/AP2A2 locus and its relevance to Alzheimer’s disease: a review. J Neuropathol Exp Neurol. 2020;79(6):568-84. https://doi.org/10.1093/jnen/nlaa024
https://doi.org/10.1093/jnen/nlaa024...
.

The GGC repeat expansion of NOTCH2NLC gene leads to neuronal intranuclear inclusion disease (NIID) and was also observed in family members affected by AD and Parkinson’s disease3939 Tian Y, Wang JL, Huang W, Zeng S, Jiao B, Liu Z, et al. Expansion of human-specific GGC repeat in neuronal intranuclear inclusion disease-related disorders. Am J Hum Genet. 2019;105(1):166-76. https://doi.org/10.1016/j.ajhg.2019.05.013
https://doi.org/10.1016/j.ajhg.2019.05.0...
. A rare nonsynonymous variant in the SHARPIN gene, p.Gly186Arg, is potentially associated with increased risk of LOAD. It leads to aberrant cellular localization of the variant protein and attenuates the activation of NF-κB, a central mediator of inflammatory and immune responses4040 Asanomi Y, Shigemizu D, Miyashita A, Mitsumori R, Mori T, Hara N, et al. A rare functional variant of SHARPIN attenuates the inflammatory response and associates with increased risk of late-onset Alzheimer’s disease. Mol Med. 2019;25(1):20. https://doi.org/10.1186/s10020-019-0090-5
https://doi.org/10.1186/s10020-019-0090-...
.

Individuals with Down syndrome (trisomy 21) developed the AD neuropathologic hallmarks after the age of 40 years, due to overexpression of APP on chromosome 21 and the resultant overproduction of β-amyloid in the brains of people’s trisomy for this gene4141 Head E, Powell D, Gold BT, Schmitt FA. Alzheimer’s disease in Down syndrome. Eur J Neurodegener Dis. 2012;1(3):353-64. PMID: 25285303.

Somatic (non-inherited) mtDNA mutations and mitochondrial dysfunction are thought to be important drivers of ageing and age-related neurodegenerative diseases such as AD4242 Srivastava S. The mitochondrial basis of aging and age-related disorders. Genes (Basel). 2017;8(12):398. https://doi.org/10.3390/genes8120398
https://doi.org/10.3390/genes8120398...
. The sequencing of OGDH, DLST, and DLD genes, encoding alpha-ketoglutarate dehydrogenase complex (αKGDHc) subunits, identified a likely pathogenic [R263H] mutation in the DLD gene associated with AD4343 Csaban D, Pentelenyi K, Toth-Bencsik R, Illes A, Grosz Z, Gezsi A, et al. The role of the rare variants in the genes encoding the alpha-ketoglutarate dehydrogenase in Alzheimer’s disease. Life (Basel). 2021;11(4):321. https://doi.org/10.3390/life11040321
https://doi.org/10.3390/life11040321...
.

Few gender-based studies have been conducted on AD disorder; nevertheless, it was found that females are at higher risk. APOE e4 females may show increased levels of AD pathology, more compromised brain network integrity, and/or accelerated longitudinal decline at a given level of AD pathology than males4444 Nebel RA, Aggarwal NT, Barnes LL, Gallagher A, Goldstein JM, Kantarci K, et al. Understanding the impact of sex and gender in Alzheimer’s disease: a call to action. Alzheimers Dement. 2018;14(9):1171-83. https://doi.org/10.1016/j.jalz.2018.04.008
https://doi.org/10.1016/j.jalz.2018.04.0...
. Greater hippocampal electroencephalograph disruption and memory impairment were seen in female ACE1 [R1279Q] KI mice, compared to males, suggesting a mechanism for higher AD risk in women4545 Cuddy LK, Prokopenko D, Cunningham EP, Brimberry R, Song P, Kirchner R, et al. Aβ-accelerated neurodegeneration caused by Alzheimer’s-associated ACE variant R1279Q is rescued by angiotensin system inhibition in mice. Sci Transl Med. 2020;12(563):eaaz2541. https://doi.org/scitranslmed.aaz2541
https://doi.org/scitranslmed.aaz2541...
.

Despite these recent advances in AD genomics, a significant part of the genetic contribution to AD remains unexplained. Further functional studies are required to examine mutation-specific expressions and understand the mechanisms by which the mutations lead to disease4646 Neuner SM, Tcw J, Goate AM. Genetic architecture of Alzheimer’s disease. Neurobiol Dis. 2020;143:104976. https://doi.org/10.1016/j.nbd.2020.104976
https://doi.org/10.1016/j.nbd.2020.10497...
. Figure 1 presents the interaction between the genes associated with Alzheimer’s disease. The functional links between these genes are identified and documented by experimental, biochemical, and expressional studies in scientific literature.

Figure 1
Interaction between the genes associated with Alzheimer’s disease.

CHALLENGES OF WHOLE-EXOME SEQUENCING AND WHOLE-GENOME SEQUENCING

The high complexity of NGS workflow and result interpretation are the major challenges encountered in WES and WGS. Most variants are inevitably detected in every individual tested and it is essential to provide a comprehensive clinical interpretation for these variants with a long time invested4747 Meienberg J, Bruggmann R, Oexle K, Matyas G. Clinical sequencing: is WGS the better WES? Hum Genet. 2016;135(3):359-62. https://doi.org/10.1007/s00439-015-1631-9
https://doi.org/10.1007/s00439-015-1631-...
. This highlights the fact that the cost for providing clinical WGS/WES is likely to remain high even as sequencing costs fall. Every step of the NGS assay requires thorough validation, therefore the sample undergoes quality checks under standard guidelines. Sequencing errors such as low depth, low alternate allele frequency, low coverage region, etc. occurring due to technical limitations, may lead to a missed variant or a false-positive result.

In genome analysis, variant calling is affected by many factors. First, polymorphic region — a region with multiple variants is scattered throughout the region and is known as a “confetti effect”. Any variant calling in this region can be challenging. Second, homopolymer repeat regions — tracts of repeated small nucleotide sequences together, which are skipped or cannot be picked up by sequencing. Third, strand bias — it occurs when reads aligned to a reference are biased towards the forward or reverse strands. It is common around exon boundaries, particularly for WES with a high chance of a false positive variant or a missed variant4848 Schwarze K, Buchanan J, Taylor JC, Wordsworth S. Are whole-exome and whole-genome sequencing approaches cost-effective? A systematic review of the literature. Genet Med. 2018;20(10):1122-30. https://doi.org/10.1038/gim.2017.247
https://doi.org/10.1038/gim.2017.247...
. Fourth, low depth of coverage — it means that the number of reads covering a region is few. Sometimes, false calls can be made by assuming polymorphic variant as a rare significant variant4949 Seaby EG, Ennis S. Challenges in the diagnosis and discovery of rare genetic disorders using contemporary sequencing technologies. Brief Funct Genomics. 2020;19(4):243-58. https://doi.org/10.1093/bfgp/elaa009
https://doi.org/10.1093/bfgp/elaa009...
. In this case, Sanger sequencing can be used to validate the variant as it tends to provide qualitative results, differently from NGS. Sanger sequencing is ideal for sequencing homogeneous samples that include one template, one gene, or one region.

Most genes referred above have 100% coverage in NGS sequencing. Few genes, such as PICALM, CD2AP and ADAM10, and CR1, are not fully covered due to segmental duplication site (pseudogenes) of polymorphic low covered regions, present in the genes (Table 3)5050 Silva R, Burch JB. Evidence that chicken CR1 elements represent a novel family of retroposons. Mol Cell Biol. 1989;9(8):3563-6. https://doi.org/10.1128/mcb.9.8.3563-3566.1989
https://doi.org/10.1128/mcb.9.8.3563-356...
.

Table 3
Average gene coverage and sequencing depth expected in sequencing for Alzheimer’s disease genes.

In a targeted-panel sequencing, the clinical importance of the genes and selective enrichment of targeted-genomic areas for NGS are the primary concerns. The selection of suitable target capture approaches and sequencing methods are crucial in yielding good quality results. This is determined by several factors such as the sample type (fresh, frozen, or formalin-fixed paraffin-embedded [FFPE]), quantity and quality of DNA or RNA routinely available5151 Yamamoto T, Shimojima K, Ondo Y, Imai K, Chong PF, Kira R, et al. Challenges in detecting genomic copy number aberrations using next-generation sequencing data and the eXome Hidden Markov Model: a clinical exome-first diagnostic approach. Hum Genome Var. 2016;3:16025. https://doi.org/10.1038/hgv.2016.25
https://doi.org/10.1038/hgv.2016.25...
.

As it is evident that every variation cannot be classified as pathogenic, a thorough validation is required before sending a final report to the patient. Research is a continuous process and functional studies on different variations can lead to upgrading or downgrading a variant classification. A revision of the variant in reports must be done timely so that clinicians can design and provide adequate treatment to patients.

Failure to state the authenticity of large deletions or duplications in genes (copy number variations) can lead to serious disorders. Therefore, cross-confirmatory tests are recommended to ascertain the CNV and its effect on the patient’s phenotype, so that treatments can be planned accordingly5252 Luthra R, Chen H, Roy-Chowdhuri S, Singh RR. Next-generation sequencing in clinical molecular diagnostics of cancer: advantages and challenges. Cancers (Basel). 2015;7(4):2023-36. https://doi.org/10.3390/cancers7040874
https://doi.org/10.3390/cancers7040874...
.

In conclusion, over the past couple of decades, high-throughput genome technologies have changed the genetic landscape of AD. NGS combined with other molecular advances, such as omics data, biochemical and functional studies, can now provide scientists with the ability to gain a comprehensive view of molecular disease pathways.

NGS assists in deducing the gain or loss of function in genes responsible for causing AD. Moreover, recent advances in NGS and its analysis have helped detect and confirm short tandem deletions and duplications. Several modifications to the current technology have been made on a day-to-day basis so that people can yield maximum benefit from NGS and help them lead a better and quality life. In the near future, both hypothesis-free (whole-genome, whole-exome) and hypothesis-driven (targeted-exome) NGS approaches will probably disentangle much of the disease genetics. Despite the few limitations stated above, it can pave the way for developing novel therapeutics and designing the right treatment on an individual level, called personalized medicine or precision medicine, which can effectively prevent or halt the progression of this devastating disease.

  • This study was conducted by the NGS laboratory, MedGenome labs Pvt. Ltd., Bangalore, India.
  • Funding: none.

REFERENCES

  • 1
    Heather JM, Chain B. The sequence of sequencers: the history of sequencing DNA. Genomics. 2016;107(1):1-8. https://doi.org/10.1016/j.ygeno.2015.11.003
    » https://doi.org/10.1016/j.ygeno.2015.11.003
  • 2
    van Dijk EL, Jaszczyszyn Y, Naquin D, Thermes C. The third revolution in sequencing technology. Trends Genet. 2018;34(9):666-81. https://doi.org/10.1016/j.tig.2018.05.008
    » https://doi.org/10.1016/j.tig.2018.05.008
  • 3
    Shademan B, Avci CB, Nikanfar M, Nourazarian A. Application of next-generation sequencing in neurodegenerative diseases: opportunities and challenges. Neuromolecular Med. 2021;23(2):225-35. https://doi.org/10.1007/s12017-020-08601-7
    » https://doi.org/10.1007/s12017-020-08601-7
  • 4
    Sun Y, Ruivenkamp CAL, Hoffer MJV, Vrijenhoek T, Kriek M, van Asperen CJ, et al. Next-generation diagnostics: gene panel, exome, or whole genome? Hum Mutat. 2015;36(6):648-55. https://doi.org/10.1002/humu.22783
    » https://doi.org/10.1002/humu.22783
  • 5
    Choudhury A, Ramsay M, Hazelhurst S, Aron S, Bardien S, Botha G, et al. Whole-genome sequencing for an enhanced understanding of genetic variation among South Africans. Nat Commun. 2017;8(1):2062. https://doi.org/10.1038/s41467-017-00663-9
    » https://doi.org/10.1038/s41467-017-00663-9
  • 6
    ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57-74. https://doi.org/10.1038/nature11247
    » https://doi.org/10.1038/nature11247
  • 7
    Jayadev S, Smith CO, Bird TD. Neurogenetics: five new things. Neurol Clin Pract. 2011;1(1):41-8. https://doi.org/10.1212/CPJ.0b013e31823c0f5f
    » https://doi.org/10.1212/CPJ.0b013e31823c0f5f
  • 8
    Gao HM, Hong JS. Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression. Trends Immunol. 2008;29(8):357-65. https://doi.org/10.1016/j.it.2008.05.002
    » https://doi.org/10.1016/j.it.2008.05.002
  • 9
    Foo JN, Liu JJ, Tan EK. Whole-genome and whole-exome sequencing in neurological diseases. Nat Rev Neurol. 2012;8(9):508-17. https://doi.org/10.1038/nrneurol.2012.148
    » https://doi.org/10.1038/nrneurol.2012.148
  • 10
    Singer J, Ruscheweyh HJ, Hofmann AL, Thurnherr T, Singer F, Toussaint NC, et al. NGS-pipe: a flexible, easily extendable and highly configurable framework for NGS analysis. Bioinformatics. 2018;34(1):107-8. https://doi.org/10.1093/bioinformatics/btx540
    » https://doi.org/10.1093/bioinformatics/btx540
  • 11
    Hess JF, Kohl TA, Kotrová M, Rönsch K, Paprotka T, Mohr V, et al. Library preparation for next generation sequencing: a review of automation strategies. Biotechnol Adv. 2020;41:107537. https://doi.org/10.1016/j.biotechadv.2020.107537
    » https://doi.org/10.1016/j.biotechadv.2020.107537
  • 12
    Thankachan A, Thomas B. A study of next generation sequencing data, workflow, application and platform comparison. Series: Materials Science and Engineering. International Conference on Recent Advancements and Effectual Researches in Engineering Science and Technology (RAEREST) 20-21 April 2018, Kerala State, India. https://doi.org/10.1088/1757-899X/396/1/012031
    » https://doi.org/10.1088/1757-899X/396/1/012031
  • 13
    Lopez JAS, González HM, Léger GC. Alzheimer’s disease. Handb Clin Neurol. 2019;167:231-55. https://doi.org/10.1016/B978-0-12-804766-8.00013-3
    » https://doi.org/10.1016/B978-0-12-804766-8.00013-3
  • 14
    Porsteinsson AP, Isaacson RS, Knox S, Sabbagh MN, Rubino I. Diagnosis of early Alzheimer’s disease: clinical practice in 2021. J Prev Alzheimers Dis. 2021;8(3):371-86. https://doi.org/10.14283/jpad.2021.23
    » https://doi.org/10.14283/jpad.2021.23
  • 15
    Li X, Feng X, Sun X, Hou N, Han F, Liu Y. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990-2019. Front Aging Neurosci. 2022;14:937486. https://doi.org/10.3389/fnagi.2022.937486
    » https://doi.org/10.3389/fnagi.2022.937486
  • 16
    Bature F, Guinn BA, Pang D, Pappas Y. Signs and symptoms preceding the diagnosis of Alzheimer’s disease: a systematic scoping review of literature from 1937 to 2016. BMJ Open. 2017;7(8):e015746. https://doi.org/10.1136/bmjopen-2016-015746
    » https://doi.org/10.1136/bmjopen-2016-015746
  • 17
    Bertram L. Next generation sequencing in Alzheimer’s disease. Methods Mol Biol. 2016;1303:281-97. https://doi.org/10.1007/978-1-4939-2627-5_17
    » https://doi.org/10.1007/978-1-4939-2627-5_17
  • 18
    Hippius H, Neundörfer G. The discovery of Alzheimer’s disease. Dialogues Clin Neurosci. 2003;5(1):101-8. https://doi.org/10.31887/DCNS.2003.5.1/hhippius
    » https://doi.org/10.31887/DCNS.2003.5.1/hhippius
  • 19
    Savage JC, Picard K, González-Ibáñez F, Tremblay ME. A brief history of microglial ultrastructure: distinctive features, phenotypes, and functions discovered over the past 60 years by electron microscopy. Front Immunol. 2018;9:803. https://doi.org/10.3389/fimmu.2018.00803
    » https://doi.org/10.3389/fimmu.2018.00803
  • 20
    Jack Jr CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14(4):535-62. https://doi.org/10.1016/j.jalz.2018.02.018
    » https://doi.org/10.1016/j.jalz.2018.02.018
  • 21
    Moloney CM, Lowe VJ, Murray ME. Visualization of neurofibrillary tangle maturity in Alzheimer’s disease: a clinicopathologic perspective for biomarker research. Alzheimers Dement. 2021;17(9):1554-74. https://doi.org/10.1002/alz.12321
    » https://doi.org/10.1002/alz.12321
  • 22
    Weller J, Budson A. Current understanding of Alzheimer’s disease diagnosis and treatment. F1000Res. 2018;7:F1000 Faculty Rev-1161. https://doi.org/10.12688/f1000research.14506.1
    » https://doi.org/10.12688/f1000research.14506.1
  • 23
    Bertram L, McQueen MB, Mullin K, Blacker D, Tanzi RE. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet. 2007;39(1):17-23. https://doi.org/10.1038/ng1934
    » https://doi.org/10.1038/ng1934
  • 24
    Zou Z, Liu C, Che C, Huang H. Clinical genetics of Alzheimer’s disease. Biomed Res Int. 2014;2014:291862. https://doi.org/10.1155/2014/291862
    » https://doi.org/10.1155/2014/291862
  • 25
    Chelban V, Breza M, Szaruga M, Vandrovcova J, Murphy D, Lee CJ, et al. Spastic paraplegia preceding PSEN1-related familial Alzheimer’s disease. Alzheimers Dement (Amst). 2021;13(1):e12186. https://doi.org/10.1002/dad2.12186
    » https://doi.org/10.1002/dad2.12186
  • 26
    Zhang Q, Sidorenko J, Couvy-Duchesne B, Marioni RE, Wright MJ, Goate AM, et.al. Risk prediction of late-onset Alzheimer’s disease implies an oligogenic architecture. Nat Commun. 2020;11(1):4799. https://doi.org/10.1038/s41467-020-18534-1
    » https://doi.org/10.1038/s41467-020-18534-1
  • 27
    Gan L, Cookson MR, Petrucelli L, La Spada AR. Converging pathways in neurodegeneration, from genetics to mechanisms. Nat Neurosci. 2018;21(10):1300-9. https://doi.org/10.1038/s41593-018-0237-7
    » https://doi.org/10.1038/s41593-018-0237-7
  • 28
    Liu CC, Liu CC, Kanekiyo T, Xu H, Bu G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol. 2013;9(2):106-18. https://doi.org/10.1038/nmeurol.2012.263
    » https://doi.org/10.1038/nmeurol.2012.263
  • 29
    Bagyinszky E, Youn YC, An SSA, Kim SY. The genetics of Alzheimer’s disease. Clin Interv Aging. 2014;9:535-51. https://doi.org/10.2147/CIA.S51571
    » https://doi.org/10.2147/CIA.S51571
  • 30
    Condello C, Yuan P, Grutzendler J. Microglia-mediated neuroprotection, TREM2, and Alzheimer’s disease: evidence from optical imaging. Biol Psychiatry. 2018;83(4):377-87. https://doi.org/10.1016/j.biopsych.2017.10.007
    » https://doi.org/10.1016/j.biopsych.2017.10.007
  • 31
    Harms M, Benitez BA, Cairns N, Cooper B, Cooper P, Mayo K, et al. C9orf72 hexanucleotide repeat expansions in clinical Alzheimer disease. JAMA Neurol. 2013;70(6):736-41. https://doi.org/10.1001/2013.jamaneurol.537
    » https://doi.org/10.1001/2013.jamaneurol.537
  • 32
    Jin SC, Pastor P, Cooper B, Cervantes S, Benitez BA, Razquin C, et al. Pooled-DNA sequencing identifies novel causative variants in PSEN1, GRN and MAPT in a clinical early-onset and familial Alzheimer’s disease Ibero-American cohort. Alzheimers Res Ther. 2012;4(4):34. https://doi.org/10.1186/alzrt137
    » https://doi.org/10.1186/alzrt137
  • 33
    Raghavan NS, Brickman AM, Andrews H, Manly JJ, Schupf N, Lantigua R, et al. Whole-exome sequencing in 20,197 persons for rare variants in Alzheimer’s disease. Ann Clin Transl Neurol. 2018;5(7):832-42. https://doi.org/10.1002/acn3.582
    » https://doi.org/10.1002/acn3.582
  • 34
    Hoogmartens J, Hens E, Engelborghs S, Vandenberghe R, De Deyn PP, Cacace R, et al. Contribution of homozygous and compound heterozygous missense mutations in VWA2 to Alzheimer’s disease. Neurobiol Aging. 2021;99:100.e17-100.e23. https://doi.org/10.1016/j.neurobiolaging.2020.09.009
    » https://doi.org/10.1016/j.neurobiolaging.2020.09.009
  • 35
    Rajabli F, Feliciano-Astacio BE, Cukier HN, Wang L, Griswold AJ, Hamilton-Nelson KL, et al. Linkage of Alzheimer disease families with Puerto Rican ancestry identifies a chromosome 9 locus. Neurobiol Aging. 2021;104:115.e1-115.e7. https://doi.org/10.1016/j.neurobiolaging.2021.02.019
    » https://doi.org/10.1016/j.neurobiolaging.2021.02.019
  • 36
    Sims R, van der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat Genet. 2017;49(9):1373-84. https://doi.org/10.1038/ng.3916
    » https://doi.org/10.1038/ng.3916
  • 37
    Cruchaga C, Karch CM, Jin SC, Benitez BA, Cai Y, Guerreiro R, et al. Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer’s disease. Nature. 2014;505(7484):550-4. https://doi.org/10.1038/nature12825
    » https://doi.org/10.1038/nature12825
  • 38
    Nelson PT, Fardo DW, Katsumata Y. The MUC6/AP2A2 locus and its relevance to Alzheimer’s disease: a review. J Neuropathol Exp Neurol. 2020;79(6):568-84. https://doi.org/10.1093/jnen/nlaa024
    » https://doi.org/10.1093/jnen/nlaa024
  • 39
    Tian Y, Wang JL, Huang W, Zeng S, Jiao B, Liu Z, et al. Expansion of human-specific GGC repeat in neuronal intranuclear inclusion disease-related disorders. Am J Hum Genet. 2019;105(1):166-76. https://doi.org/10.1016/j.ajhg.2019.05.013
    » https://doi.org/10.1016/j.ajhg.2019.05.013
  • 40
    Asanomi Y, Shigemizu D, Miyashita A, Mitsumori R, Mori T, Hara N, et al. A rare functional variant of SHARPIN attenuates the inflammatory response and associates with increased risk of late-onset Alzheimer’s disease. Mol Med. 2019;25(1):20. https://doi.org/10.1186/s10020-019-0090-5
    » https://doi.org/10.1186/s10020-019-0090-5
  • 41
    Head E, Powell D, Gold BT, Schmitt FA. Alzheimer’s disease in Down syndrome. Eur J Neurodegener Dis. 2012;1(3):353-64. PMID: 25285303
  • 42
    Srivastava S. The mitochondrial basis of aging and age-related disorders. Genes (Basel). 2017;8(12):398. https://doi.org/10.3390/genes8120398
    » https://doi.org/10.3390/genes8120398
  • 43
    Csaban D, Pentelenyi K, Toth-Bencsik R, Illes A, Grosz Z, Gezsi A, et al. The role of the rare variants in the genes encoding the alpha-ketoglutarate dehydrogenase in Alzheimer’s disease. Life (Basel). 2021;11(4):321. https://doi.org/10.3390/life11040321
    » https://doi.org/10.3390/life11040321
  • 44
    Nebel RA, Aggarwal NT, Barnes LL, Gallagher A, Goldstein JM, Kantarci K, et al. Understanding the impact of sex and gender in Alzheimer’s disease: a call to action. Alzheimers Dement. 2018;14(9):1171-83. https://doi.org/10.1016/j.jalz.2018.04.008
    » https://doi.org/10.1016/j.jalz.2018.04.008
  • 45
    Cuddy LK, Prokopenko D, Cunningham EP, Brimberry R, Song P, Kirchner R, et al. Aβ-accelerated neurodegeneration caused by Alzheimer’s-associated ACE variant R1279Q is rescued by angiotensin system inhibition in mice. Sci Transl Med. 2020;12(563):eaaz2541. https://doi.org/scitranslmed.aaz2541
    » https://doi.org/scitranslmed.aaz2541
  • 46
    Neuner SM, Tcw J, Goate AM. Genetic architecture of Alzheimer’s disease. Neurobiol Dis. 2020;143:104976. https://doi.org/10.1016/j.nbd.2020.104976
    » https://doi.org/10.1016/j.nbd.2020.104976
  • 47
    Meienberg J, Bruggmann R, Oexle K, Matyas G. Clinical sequencing: is WGS the better WES? Hum Genet. 2016;135(3):359-62. https://doi.org/10.1007/s00439-015-1631-9
    » https://doi.org/10.1007/s00439-015-1631-9
  • 48
    Schwarze K, Buchanan J, Taylor JC, Wordsworth S. Are whole-exome and whole-genome sequencing approaches cost-effective? A systematic review of the literature. Genet Med. 2018;20(10):1122-30. https://doi.org/10.1038/gim.2017.247
    » https://doi.org/10.1038/gim.2017.247
  • 49
    Seaby EG, Ennis S. Challenges in the diagnosis and discovery of rare genetic disorders using contemporary sequencing technologies. Brief Funct Genomics. 2020;19(4):243-58. https://doi.org/10.1093/bfgp/elaa009
    » https://doi.org/10.1093/bfgp/elaa009
  • 50
    Silva R, Burch JB. Evidence that chicken CR1 elements represent a novel family of retroposons. Mol Cell Biol. 1989;9(8):3563-6. https://doi.org/10.1128/mcb.9.8.3563-3566.1989
    » https://doi.org/10.1128/mcb.9.8.3563-3566.1989
  • 51
    Yamamoto T, Shimojima K, Ondo Y, Imai K, Chong PF, Kira R, et al. Challenges in detecting genomic copy number aberrations using next-generation sequencing data and the eXome Hidden Markov Model: a clinical exome-first diagnostic approach. Hum Genome Var. 2016;3:16025. https://doi.org/10.1038/hgv.2016.25
    » https://doi.org/10.1038/hgv.2016.25
  • 52
    Luthra R, Chen H, Roy-Chowdhuri S, Singh RR. Next-generation sequencing in clinical molecular diagnostics of cancer: advantages and challenges. Cancers (Basel). 2015;7(4):2023-36. https://doi.org/10.3390/cancers7040874
    » https://doi.org/10.3390/cancers7040874

Publication Dates

  • Publication in this collection
    07 Aug 2023
  • Date of issue
    2023

History

  • Received
    10 Apr 2022
  • Reviewed
    10 Apr 2023
  • Accepted
    17 May 2023
Academia Brasileira de Neurologia, Departamento de Neurologia Cognitiva e Envelhecimento R. Vergueiro, 1353 sl.1404 - Ed. Top Towers Offices, Torre Norte, São Paulo, SP, Brazil, CEP 04101-000, Tel.: +55 11 5084-9463 | +55 11 5083-3876 - São Paulo - SP - Brazil
E-mail: revistadementia@abneuro.org.br | demneuropsy@uol.com.br