Page 109 - Read Online
P. 109

Page 243                Ratnapriya. J Transl Genet Genom 2022;6:240-256  https://dx.doi.org/10.20517/jtgg.2021.54

               STEPS TO CONNECT GWAS DISCOVERY TO BIOLOGY
               GWAS unquestionably established a strong genetic component of AMD and provided a broad framework
               for elucidating the contribution of genetic variants to AMD. It allowed a hypotheses-free discovery of
               disease variants that revealed novel disease biology and provided a wealth of clues for biological
               experimentation, risk prediction, disease progression, and biomarker and therapeutic target discovery
               [Figure 1]. However, we must also acknowledge that progress in translating genetic findings into
               understanding disease mechanisms and new cures and treatments for AMD has been slow to arrive. There
               are no biomarkers for predicting the disease progression to either GA or CNV. Unfortunately, targeting
               GWAS implicated pathways has not been effective in preventing/slowing disease progression or treatment
                                                                                      [44]
               as many clinical trials that relied on these hypotheses have failed to ameliorate AMD .

               Drug targets with human genetic evidence of disease association are twice as likely to succeed as approved
                    [45]
               drugs . Thus, investments in the functional follow-up of genomic findings are likely to be beneficial for
               AMD drug development. However, several factors have made it difficult to bridge the gap between the
               statistical associations of genetic variations and a functional understanding of the biology underlying AMD
               risk. First, GWAS variants are not causal. The causative SNP may lie anywhere within the linkage
               disequilibrium (LD) block surrounding the associated SNP that can span over 100 kb and often contain over
               a thousand individual SNPs. Second, a majority of associated variants reside in non-coding regions of the
               genome, thus underlying causative genes or pathological mechanisms are not immediately obvious. Some of
               the AMD-specific challenges are also worth noting. For example, modeling of AMD presents a substantial
               challenge . AMD causes vision loss because of degeneration of photoreceptors in the central region of the
                       [46]
                                                                                                       [47]
               retina called the macula, which is a primate-specific structure shared only by humans, monkeys and apes .
               Thus, mouse models fall short of recapitulating many aspects of AMD. Similarly, there is a dearth of cell
               lines for ocular studies, and transformed cell lines such as ARPE-19 do not express key phenotypic features
               of human RPE . Finally, we have a limited understanding of molecular and genetic underpinnings in the
                            [48]
               early and intermediate stages of the disease.

               There are four key steps in elucidating the disease mechanisms underlying AMD risk variants: (1)
               identification of causal variant(s) and target gene(s) at the associated loci; (2) annotating variants and genes
               at tissues and single-cell resolution; (3) identifying molecular and cellular pathways associated with the
               disease; and (4) developing appropriate disease models to infer disease mechanisms. As a vast majority of
               GWAS variants fall into non-coding regions and often overlap enhancers, promoters and open-chromatin
               regions, gene expression regulation is emerging as a dominant mechanism in mediating disease risk. Thus,
               here we will be focusing on various ways gene expression studies can be leveraged to understand the risk
               variant-mediated disease causation.


               TRANSCRIPTOME STUDIES IN DISEASE-RELEVANT TISSUES AND CELL TYPES
               GWAS have established that > 90% of the risk variants associated with complex diseases including AMD
               reside in the non-coding regions that are not likely to directly affect the coding region of the gene. This
               brought attention to the annotation of the non-coding genome to understand its function. Enrichment of
               GWAS variants within regulatory DNA marked by deoxyribonuclease I (DNase I) hypersensitive sites
                                                                                                       [49]
               (DHSs) led the focus on studying the gene expression regulation in the context of GWAS variants .
               Additionally, these risk variants often disrupt binding sites for transcription factors (TFs), and these variable
               TF-interaction are believed to be the primary driver of phenotypic variation . Additionally, cell context is a
                                                                               [50]
               key determinant of gene regulation. Thus, a comprehensive understanding of global transcriptome
               regulation in disease-relevant cells and tissues represents the first logical step in functional understanding of
               GWAS findings. Towards this goal, Genotype-Tissue Expression (GTEx) project was initiated to establish a
   104   105   106   107   108   109   110   111   112   113   114