Complex traits

From Wikipedia the free encyclopedia

The size of a tomato is one example of a complex trait.

Complex traits are phenotypes that are controlled by two or more genes and do not follow Mendel’s Law of Dominance. They may have a range of expression which is typically continuous. Both environmental and genetic factors often impact the variation in expression. Human height is a continuous trait meaning that there is a wide range of heights. There are an estimated 50 genes that affect the height of a human. Environmental factors, like nutrition, also play a role in a human’s height. Other examples of complex traits include: crop yield, plant color, and many diseases including diabetes and Parkinson's disease. One major goal of genetic research today is to better understand the molecular mechanisms through which genetic variants act to influence complex traits. Complex Traits are also known as polygenic traits and multigenic traits. [1][2]

The existence of complex traits, which are far more common than Mendelian traits, represented a significant challenge to the acceptance of Mendel’s work. Modern understanding has 3 categories of complex traits: quantitative, meristic, and threshold. These traits have been studied on a small scale with observational techniques like twin studies. They are also studied with statistical techniques like quantitative trait loci(QTL) mapping, and genome-wide association studies(GWAS) on a large scale. The overall goal of figuring out how genes interact with each other and the environment and how those interactions can lead to variation in a trait is called genetic architecture.

History[edit]

When Mendel's work on inheritance was rediscovered in 1900, scientists debated whether Mendel's laws could account for the continuous variation observed for many traits.[citation needed] One group known as the biometricians argued that continuous traits such as height were largely heritable, but could not be explained by the inheritance of single Mendelian genetic factors. Work published by Ronald Fisher in 1919 mostly resolved debate by demonstrating that the variation in continuous traits could be accounted for if multiple such factors contributed additively to each trait.[1] However, the number of genes involved in such traits remained undetermined; until recently, genetic loci were expected to have moderate effect sizes and each explain several percent of heritability.[3] After the conclusion of the Human Genome Project in 2001, it seemed that the sequencing and mapping of many individuals would soon allow for a complete understanding of traits' genetic architectures. However, variants discovered through genome-wide association studies (GWASs) accounted for only a small percentage of predicted heritability; for example, while height is estimated to be 80-90% heritable, early studies only identified variants accounting for 5% of this heritability.[4] Later research showed that most missing heritability could be accounted for by common variants missed by GWASs because their effect sizes fell below significance thresholds; a smaller percentage is accounted for by rare variants with larger effect sizes, although in certain traits such as autism, rare variants play a more dominant role.[5][6][7] While many genetic factors involved in complex traits have been identified, determining their specific contributions to phenotypes—specifically, the molecular mechanisms through which they act—remains a major challenge.[8]

Types of Complex Traits[edit]

Quantitative Traits[edit]

Quantitative Traits have phenotypes that are expressed on continuous ranges.[9] They have many different genes that impact the phenotype, with differing effect sizes.[10] Many of these traits are some what heritable. For example, height is estimated to be 60-80% heritable; however, other quantitative traits have varying heritability.[11]

Meristic Traits[edit]

Meristic traits have phenotypes that are described by whole numbers. An example is the rate chickens lay eggs. A chicken can lay one, two, or five eggs a week, but never half an egg.[9] The environment can also impact expression, as chickens won’t lay as many eggs depending on the time of year.[12]

Threshold Traits[edit]

Threshold traits have phenotypes that have limited expressions (usually two). It is a complex trait because multiple genetic and environmental factors impact the phenotype.[13][14] The phenotype before the threshold is referred to as normal or absent, and after the threshold as lethal or present. These traits are often examined in a medical context, because many diseases exhibit this pattern or similar.[9] An example of this is type 2 diabetes, the phenotype is either normal/healthy or lethal/diseased.[15]

Methods for Finding Complex Traits[edit]

Twin Studies[edit]

Twin Studies is an observational test using monozygotic twins and dizygotic twins, preferably same sex. They are used to figure out the environmental influence on complex traits. Monozygotic twins in particular are estimated to share 100% of their DNA with each other so any phenotypic differences should be caused by environmental influences.[2]

QTL mapping[edit]

A quantitative trait locus (QTL) is a section of the genome associated with variation in a quantitative, or complex, trait. To identify QTLs, QTL mapping is performed on individuals with differing genotypes. First, mapping involves either full-genome sequencing or the genotyping of many marker loci throughout the genome; then, phenotypes of interest are measured. For example, the expression levels of different genes in the genome is one commonly-measured phenotype (the associated loci are called eQTLs). At each locus, individuals are grouped by their genotype, and statistical tests are performed to determine whether measured trait values for one group differ significantly from the overall mean for all groups. Identified loci may not be QTLs themselves, but are likely in linkage disequilibrium—and therefore strongly associate—with the loci actually influencing the trait.

GWAS[edit]

A Genome-Wide Association Study (GWAS) is a technique used to find gene variants linked to complex traits. A GWAS is done with populations that mate randomly because all the genetic variants are tested at once. Then researchers can compare the different alleles at a locus. It is similar to QTL mapping.[16] The most common set-up for a GWAS is a case study which creates two populations one with the trait we are looking at and one without the trait. With the two populations researchers will map every subject's genome and compare them to find different variance in the SNPs between the two populations.[citation needed] Both populations should have similar environmental backgrounds. GWAS is only looking at the DNA and does not include differences that would be caused by environmental factors.[2]

A manhattan plot showing genome-association with microcirculation.

Statistical test, such as a chi squared is used to find if there is association with the trait and each of the SNPs tested. The statistical test produces a p-value which the researcher will use to conclude if the SNP is significant. This p-value cut off can range from being a higher number or a lower number at the researcher's discretion. The data can then be visualized in a Manhattan plot which takes the -log (p-value) so all the significant SNPs are at the top of the graph.[17][18]

Genetic architecture[edit]

Genetic architecture is an overall explanation of all the genetic factors that play a role in a complex trait and exists as the core foundation of quantitative genetics. With the use of mathematical models and statistical analysis, like GWAS, researchers can determine the number of genes affecting a trait as well as the level of influence each gene has on the trait. This is not always easy as the architecture of one trait can be different between two separate populations of the same species.[19] This can be due to the fact that both populations live in different environments. Differing environments can lead to different interactions between genes and the environment, changing the architecture of both populations.[20]

Recently, with rapid increases in available genetic data, researchers have begun to characterize the genetic architecture of complex traits better. One surprise has been the observation that most loci identified in GWASs are found in noncoding regions of the genome; therefore, instead of directly altering protein sequences, such variants likely affect gene regulation.[21] To understand the precise effects of these variants, QTL mapping has been employed to examine data from each step of gene regulation; for example, mapping RNA-sequencing data can help determine the effects of variants on mRNA expression levels, which then presumably affect the numbers of proteins translated. A comprehensive analysis of QTLs involved in various regulatory steps—promotor activity, transcription rates, mRNA expression levels, translation levels, and protein expression levels—showed that high proportions of QTLs are shared, indicating that regulation behaves as a “sequential ordered cascade” with variants affecting all levels of regulation.[22] Many of these variants act by affecting transcription factor binding and other processes that alter chromatin function—steps which occur before and during RNA transcription.[22]

To determine the functional consequences of these variants, researchers have largely focused on identifying key genes, pathways, and processes that drive complex trait behavior; an inherent assumption has been that the most statistically significant variants have the greatest impact on traits because they act by affecting these key drivers.[8][23] For example, one study hypothesizes that there exist rate-limiting genes pivotal to the function of gene regulatory networks.[24] Others studies have identified the functional impacts of key genes and mutations on disorders, including autism and schizophrenia.[7][25] However, a 2017 analysis by Boyle et al. argues that while genes which directly impact complex traits do exist, regulatory networks are so interconnected that any expressed gene affects the functions of these "core" genes; this idea is called the "omnigenic" hypothesis.[8] While these "peripheral" genes each have small effects, their combined impact far exceeds the contributions of core genes themselves. To support the hypothesis that core genes play a smaller than expected role, the authors describe three main observations: the heritability for complex traits is spread broadly, often uniformly, across the genome; genetic effects do not appear to be mediated by cell-type specific function; and genes in the relevant functional categories only modestly contribute more to heritability than other genes.[8] One alternative to the omnigenic hypothesis is the idea that peripheral genes act not by altering core genes but by altering cellular states, such as the speed of cell division or hormone response.[26][27]

References[edit]

  1. ^ a b Fisher, R. A. (1919). "XV.—The Correlation between Relatives on the Supposition of Mendelian Inheritance". Earth and Environmental Science Transactions of the Royal Society of Edinburgh. 52 (2): 399–433. doi:10.1017/S0080456800012163. S2CID 181213898.
  2. ^ a b c Rowe, Suzanne J.; Tenesa, Albert (2012). "Human Complex Trait Genetics: Lifting the Lid of the Genomics Toolbox - from Pathways to Prediction". Current Genomics. 13 (3): 213–224. doi:10.2174/138920212800543101. PMC 3382276. PMID 23115523.
  3. ^ Gibson G (January 2012). "Rare and common variants: twenty arguments". Nature Reviews. Genetics. 13 (2): 135–45. doi:10.1038/nrg3118. PMC 4408201. PMID 22251874.
  4. ^ Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, et al. (October 2009). "Finding the missing heritability of complex diseases". Nature. 461 (7265): 747–53. Bibcode:2009Natur.461..747M. doi:10.1038/nature08494. PMC 2831613. PMID 19812666.
  5. ^ Shi H, Kichaev G, Pasaniuc B (July 2016). "Contrasting the Genetic Architecture of 30 Complex Traits from Summary Association Data". American Journal of Human Genetics. 99 (1): 139–53. doi:10.1016/j.ajhg.2016.05.013. PMC 5005444. PMID 27346688.
  6. ^ Marouli E, Graff M, Medina-Gomez C, Lo KS, Wood AR, Kjaer TR, et al. (February 2017). "Rare and low-frequency coding variants alter human adult height". Nature. 542 (7640): 186–190. Bibcode:2017Natur.542..186M. doi:10.1038/nature21039. PMC 5302847. PMID 28146470.
  7. ^ a b Krumm N, Turner TN, Baker C, Vives L, Mohajeri K, Witherspoon K, Raja A, Coe BP, Stessman HA, He ZX, Leal SM, Bernier R, Eichler EE (June 2015). "Excess of rare, inherited truncating mutations in autism". Nature Genetics. 47 (6): 582–8. doi:10.1038/ng.3303. PMC 4449286. PMID 25961944.
  8. ^ a b c d Boyle EA, Li YI, Pritchard JK (June 2017). "An Expanded View of Complex Traits: From Polygenic to Omnigenic". Cell. 169 (7): 1177–1186. doi:10.1016/j.cell.2017.05.038. PMC 5536862. PMID 28622505.
  9. ^ a b c Klug, William S. (2012). Concepts of Genetics. Pearson Education. ISBN 978-0-321-72412-0.[page needed]
  10. ^ DiCOTATO, ALLESSANDRA (October 14, 2022). "Scientists Uncover Nearly All Genetic Variants Linked to Height". Harvard Medical School. Retrieved May 10, 2024.
  11. ^ "Is height determined by genetics?: MedlinePlus Genetics". medlineplus.gov. Retrieved 2024-05-10.
  12. ^ EZMFrdmHtchy (2022-12-05). "How to Keep Chickens Laying Eggs in the Winter | Freedom Ranger Hatchery". Retrieved 2024-05-10.
  13. ^ "threshold trait / threshold traits". Scitable.
  14. ^ Pierce, Benjamin A. (2012). Genetics: a conceptual approach (4 ed.). Basingstoke: Palgrave. p. 662. ISBN 978-1-4292-3252-4.
  15. ^ Rosales-Gómez, Roberto Carlos; López-Jiménez, José de Jesús; Núñez-Reveles, Nelly Yazmine; González-Santiago, Ana Elizabeth; Ramírez-García, Sergio Alberto (2010). "Nefropatía por diabetes mellitus tipo 2: un rasgo multifactorial con umbral y su mapa mórbido cromosómico" [Type 2 diabetes nephropathy: a thresholds complex trait and chromosomal morbid map]. Revista Medica del Instituto Mexicano del Seguro Social (in Spanish). 48 (5): 521–530. PMID 21205501.
  16. ^ Griffiths, Anthony J. F.; Wessler, Susan R.; Carroll, Sean B.; Doebley, John (2015). An Introduction to Genetic Analysis. Macmillan Learning. ISBN 978-1-4641-0948-5.[page needed]
  17. ^ "Wayback Machine" (PDF). Archived from the original (PDF) on 2018-06-29. Retrieved 2024-05-10.
  18. ^ Feldman, Igor; Rzhetsky, Andrey; Vitkup, Dennis (18 March 2008). "Network properties of genes harboring inherited disease mutations". Proceedings of the National Academy of Sciences. 105 (11): 4323–4328. Bibcode:2008PNAS..105.4323F. doi:10.1073/pnas.0701722105. PMC 2393821. PMID 18326631.
  19. ^ Griffiths, Anthony J. F.; Wessler, Susan R.; Carroll, Sean B.; Doebley, John (2015). An Introduction to Genetic Analysis. Macmillan Learning. ISBN 978-1-4641-0948-5.[page needed]
  20. ^ Timpson, Nicholas J.; Greenwood, Celia M. T.; Soranzo, Nicole; Lawson, Daniel J.; Richards, J. Brent (February 2018). "Genetic architecture: the shape of the genetic contribution to human traits and disease". Nature Reviews Genetics. 19 (2): 110–124. doi:10.1038/nrg.2017.101. PMID 29225335.
  21. ^ Frazer KA, Murray SS, Schork NJ, Topol EJ (April 2009). "Human genetic variation and its contribution to complex traits". Nature Reviews. Genetics. 10 (4): 241–51. doi:10.1038/nrg2554. PMID 19293820. S2CID 19987352.
  22. ^ a b Li YI, van de Geijn B, Raj A, Knowles DA, Petti AA, Golan D, Gilad Y, Pritchard JK (April 2016). "RNA splicing is a primary link between genetic variation and disease". Science. 352 (6285): 600–4. Bibcode:2016Sci...352..600L. doi:10.1126/science.aad9417. PMC 5182069. PMID 27126046.
  23. ^ Callaway E (2017-06-15). "New concerns raised over value of genome-wide disease studies". Nature. 546 (7659): 463. doi:10.1038/nature.2017.22152.
  24. ^ Chakravarti A, Turner TN (June 2016). "Revealing rate-limiting steps in complex disease biology: The crucial importance of studying rare, extreme-phenotype families". BioEssays. 38 (6): 578–86. doi:10.1002/bies.201500203. PMID 27062178. S2CID 3813041.
  25. ^ Sekar A, Bialas AR, de Rivera H, Davis A, Hammond TR, Kamitaki N, Tooley K, Presumey J, Baum M, Van Doren V, Genovese G, Rose SA, Handsaker RE, Daly MJ, Carroll MC, Stevens B, McCarroll SA (February 2016). "Schizophrenia risk from complex variation of complement component 4". Nature. 530 (7589): 177–83. Bibcode:2016Natur.530..177.. doi:10.1038/nature16549. PMC 4752392. PMID 26814963.
  26. ^ Preininger M, Arafat D, Kim J, Nath AP, Idaghdour Y, Brigham KL, Gibson G (2013-03-14). "Blood-informative transcripts define nine common axes of peripheral blood gene expression". PLOS Genetics. 9 (3): e1003362. doi:10.1371/journal.pgen.1003362. PMC 3597511. PMID 23516379.
  27. ^ He X (October 2017). "Comment on: An Expanded View of Complex Traits: From Polygenic to Omnigenic". Journal of Psychiatry and Brain Science. 2 (5). doi:10.20900/jpbs.20170014s2.