Contemporary, yeast-based approaches to understanding human genetic variation

https://doi.org/10.1016/j.gde.2013.10.001Get rights and content

Determining how genetic variation contributes to human health and disease is a critical challenge. As one of the most genetically tractable model organisms, yeast has played a central role in meeting this challenge. The advent of new technologies, including high-throughput DNA sequencing and synthesis, proteomics, and computational methods, has vastly increased the power of yeast-based approaches to determine the consequences of human genetic variation. Recent successes include systematic exploration of the effects of gene dosage, large-scale analysis of the effect of coding variation on gene function, and the use of humanized yeast to model disease. By virtue of its manipulability, small genome size, and genetic tractability, yeast is poised to help us understand human genetic variation.

Introduction

With acceleration of sequencing technologies, many human genomes are becoming available from patients, tumors, and thousands of individuals from diverse populations. In parallel, linkage mapping, genome-wide association strategies, and analyses of de novo mutations are rapidly linking genomic regions to phenotypes including disease susceptibility. However, defining which genetic variants are causative for phenotype has become rate-limiting. Furthermore, the abundance of rare variation means that sequencing more genomes is unlikely to solve this problem (e.g. [1, 2]). We propose that new technologies such as high-throughput DNA sequencing, proteomics, and computational approaches can empower model organism genetics to fill this gap by enabling high-throughput, generic, genome-scale functional assays for characterizing variation in the human genome. Yeast, especially the budding yeast S. cerevisiae, is uniquely suited to this task because of its versatility, small genome size, and powerful array of existing tools (reviewed in [3]). Methods for understanding the consequences of human variation using yeast fall into three broad categories: 1. systematic analysis of gene dosage; 2. recreation of human variants in their yeast orthologs; and 3. cross-species complementation and heterologous expression. In addition to enabling direct measurement of the consequences of specific genetic variants, work in yeast and other model organisms will be necessary for understanding the essential underlying biology. These larger biological questions include the distribution of effect sizes of genetic variants, the contribution of genetic modifiers and the role of epistasis more generally, and, of course, the fundamental molecular mechanisms by which genes and their variants act.

Section snippets

Systematic analysis of gene dosage

Yeast is easily amenable to purposeful manipulation of gene dosage, most frequently via loss of function but increasingly by overexpression as well. Examining the resulting phenotypes can reveal the function of the element whose dosage is changed (Figure 1a). When specific phenotypes are shared, connections between yeast and human can be relatively easy to recognize. Famously, work in yeast correctly predicted the role of the human mismatch repair genes hPMS1, hMLH1, and hMSH2 in hereditary

Recreation of genetic variants in orthologs

Complete loss-of-function alleles comprise a minority of the relevant genetic variation in humans and other organisms. Most genes have many alleles ranging from complete loss of function to subtle alterations in function. For genomic regions with significant conservation, human variants can be tested for function by making homologous mutations in their yeast orthologs (Figure 1b). For example, MSH2 alleles associated with hereditary colon cancer were systematically evaluated in yeast, where

Cross-species complementation and heterologous expression

Despite the value of using orthologous genes, conservation-based inference of mutation effects can be fraught [35]. In some cases, direct testing of variants in their native gene context might be more desirable. In yeast, this has been attempted in two ways: cross-species complementation and heterologous expression. Cross-species complementation is simply the ability of human genes to rescue an orthologous loss-of-function mutation in another organism (Figure 1c). The conservation of core cell

Limitations of model organism approaches

Of course, all model systems have their downsides. Approaches in yeast might be most productively viewed as a method for intelligently prioritizing experiments to be done in more complicated and expensive mammalian models. An obvious caveat of evaluating human alleles in other organisms is that some genes will not be equivalently functional outside their native context of a human cell. Furthermore, some variants may disrupt interactions with proteins not present or too diverged in other

Conclusion

We have argued that yeast is an ideal model organism in which to address the consequences of human genomic variation. We have reviewed the key approaches that have already allowed huge progress in understanding human and yeast genetic perturbations, ranging from single point mutations to entire extra chromosomes. Additionally, the genetics underlying disease is sometimes so poorly understood that model organisms are needed just to define these basics (reviewed in [61]). For example, complex

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

MD is a Rita Allen Foundation Scholar and a Canadian Institute for Advanced Research Fellow. She is supported by grants from the National Institute of General Medical Sciences (P41 GM103533, R01GM094306 and R01GM101091) from the National Institutes of Health.

References (66)

  • O. Zuk et al.

    The mystery of missing heritability: genetic interactions create phantom heritability

    Proc Natl Acad Sci USA

    (2012)
  • M.R. Nelson et al.

    An abundance of rare functional variants in 202 drug target genes sequenced in 14,002 people

    Science

    (2012)
  • J.A. Tennessen et al.

    Evolution and functional impact of rare coding variation from deep sequencing of human exomes

    Science

    (2012)
  • D. Botstein et al.

    Yeast: an experimental organism for 21st Century biology

    Genetics

    (2011)
  • M. Strand et al.

    Destabilization of tracts of simple repetitive DNA in yeast by mutations affecting DNA mismatch repair

    Nature

    (1993)
  • F. Perocchi et al.

    Systematic screens for human disease genes, from yeast to human and back

    Mol Biosyst

    (2008)
  • L. Steinmetz et al.

    Systematic screen for human disease genes in yeast

    Nat Genet

    (2002)
  • G. Giaever et al.

    Functional profiling of the Saccharomyces cerevisiae genome

    Nature

    (2002)
  • E.A. Winzeler et al.

    Functional characterization of the S. cerevisiae genome by gene deletion and parallel analysis

    Science

    (1999)
  • C.H. Ho et al.

    A molecular barcoded yeast ORF library enables mode-of-action analysis of bioactive compounds

    Nat Biotechnol

    (2009)
  • D.C. Hess et al.

    Computationally driven, quantitative experiments discover genes required for mitochondrial biogenesis

    PLoS Genet

    (2009)
  • M.E. Hillenmeyer et al.

    The chemical genomic portrait of yeast: uncovering a phenotype for all genes

    Science

    (2008)
  • A. Deutschbauer et al.

    Parallel phenotypic analysis of sporulation and postgermination growth in Saccharomyces cerevisiae

    Proc Natl Acad Sci USA

    (2002)
  • E.J. Dean et al.

    Pervasive and persistent redundancy among duplicated genes in yeast

    PLoS Genet

    (2008)
  • G. Musso et al.

    The extensive and condition-dependent nature of epistasis among whole-genome duplicates in yeast

    Genome Res

    (2008)
  • A. Deutschbauer et al.

    Mechanisms of haploinsufficiency revealed by genome-wide profiling in yeast

    Genetics

    (2005)
  • D. Delneri et al.

    Identification and characterization of high-flux-control genes of yeast through competition analyses in continuous cultures

    Nat Genet

    (2008)
  • M. de Clare et al.

    Copy-number variation of cancer-gene orthologs is sufficient to induce cancer-like symptoms in Saccharomyces cerevisiae

    BMC Biol

    (2013)
  • M. Costanzo et al.

    The genetic landscape of a cell

    Science

    (2010)
  • D.M. van Pel et al.

    Saccharomyces cerevisiae genetics predicts candidate therapeutic genetic interactions at the mammalian replication fork

    G3

    (2013)
  • J.L. McLellan et al.

    Synthetic lethality of cohesins with PARPs and replication fork mediators

    PLoS Genet

    (2012)
  • B. Haarer et al.

    Modeling complex genetic interactions in a simple eukaryotic genome: actin displays a rich spectrum of complex haploinsufficiencies

    Genes Dev

    (2007)
  • B. Haarer et al.

    Novel interactions between actin and the proteasome revealed by complex haploinsufficiency

    PLoS Genet

    (2011)
  • Cited by (27)

    • The composition and function of the striatin-interacting phosphatases and kinases (STRIPAK) complex in fungi

      2016, Fungal Genetics and Biology
      Citation Excerpt :

      Therefore, investigating complex eukaryotic signaling pathways in microbes when functions still have to be determined can be highly relevant for higher eukaryotes, including humans. It was shown for various fungi such as budding and fission yeast, and various filamentous fungi that they are extremely useful tools for gaining mechanistic insights into cellular and developmental processes relevant for eukaryotes in general (Dunham and Fowler, 2013; Simon and Plattner, 2014). Here, we focus on the recently identified eukaryotic STRIPAK complex, which was characterized by mass spectrometry in mammals (Goudreault et al., 2009).

    • Applications of comparative evolution to human disease genetics

      2015, Current Opinion in Genetics and Development
      Citation Excerpt :

      Platforms in yeast, plants, worms, and fruit flies now exist to record quantitative data on multiple levels, including morphology, metabolism, transcription, and translation [16•,18–21]. Combining these measurements make it possible not only to uncover the biology of model systems, but also to screen human genes and candidate disease alleles in a model system background, a method that Jasper Rine and colleagues termed ‘surrogate genetics’ [22,23•]. Although model organisms will remain a mainstay of human disease genetics, the advent of these novel molecular tools has raised the possibility of high-throughput screens of genotype/phenotype relations in any organism of interest, blurring the lines between model and non-model organism.

    • The laboratory domestication of Caenorhabditis elegans

      2015, Trends in Genetics
      Citation Excerpt :

      Many different species have these attributes. For example, the single-cell eukaryote Saccharomyces cerevisiae (baker's yeast) is immensely powerful as a genetic model organism for conserved cellular processes [1] and for quantitative genetics using large populations [2]. The fruit fly Drosophila melanogaster contributed extensively to our understanding of signal-transduction pathways and developmental patterning [3].

    • Yeast cells expressing the human mitochondrial dna polymerase reveal correlations between polymerase fidelity and human disease progression

      2014, Journal of Biological Chemistry
      Citation Excerpt :

      Thus, despite evolutionary relationships between yeast and humans, one might question whether the human mtDNA polymerase could provide functional substitution for the yeast polymerase. The “humanized” yeast system, with the yeast genes replaced by their human orthologs, has been developed to rapidly examine the pathology of human genetic variation, thanks to the powerful genetic tools available for yeast (32–35). Here, we begin addressing important questions by developing “humanized” yeast expressing human mitochondrial DNA polymerase genes with wild-type and mutant alleles.

    View all citing articles on Scopus
    View full text