The generation of 1000s of patient exomes and genomes, while accelerating diagnoses, has also highlighted the complexity of previously-considered ?simple? traits. Numerous examples are now reported of patients with deleterious alleles in multiple genes. In some, the phenotype reflects an amalgam of two disorders; in others, it is the product of genetic interactions. Moreover, recent exome- or panel-based resequencing of groups of genes known to cause a host of dominant or recessive disorders are beginning to report an enrichment of rare variants in patients, highlighting the concept of mutational burden. Bardet-Biedl syndrome (BBS), a founding ciliopathy, has been a model for studying these phenomena and for beginning to understand the contribution of such alleles to non-penetrance and variable expressivity. This is because: (a) the majority of the recessive burden in BBS is now known, with ~80% of BBS patients harboring recessive mutations in 22 genes; (b) most BBS proteins are necessary for cilia structure/function and assemble into defined complexes; and (c) we and others have developed quantitative in vitro and in vivo tools to assess the total functional output of the cilium; to establish the effect of variants; and to measure genetic interactions. This Renewal focuses on three themes. First, we will capitalize on extensive genetic and functional data from previous cycles to build a comprehensive map of mutational distribution in a biological module. Through the rigorous analysis of two independent BBS patient cohorts, we have observed a 2.5-fold increase of rare variants in known BBS genes beyond the recessive driver. This variation is not distributed randomly but intimates an interaction between mutations that map to different macromolecular complexes. Using established, mouse models of BBS, we will ask how these interactions might potentiate or exacerbate discrete BBS endophenotypes. Second, we will take advantage of recent observations in humans and mouse models of ciliopathies to dissect the role of cis- and -trans acting genetic modifiers. Focusing on TTC21B/IFT139, a gene that contributes causal and modifying mutations across the ciliopathy disease spectrum, we will test the hypothesis that mutations in that locus contribute to the development of renal disease in BBS, by leveraging extensive data from zebrafish models and testing the paradigm in the mouse. In parallel, using a combination of computational and biological tools, will ask whether discrete point mutations in that locus can account for the variable activity of the disease-causing allele p.P209L, a hypomorph in human but a null allele in the mouse. Finally, grounded on recent observations derived from a genome-wide genetic suppressor screen that augmentation of the proteasome ameliorates BBS in vitro and in vivo, we will employ our recently-developed proteasome sensor screening paradigm to perform a small molecule screen for new therapeutic leads. These studies will provide a comprehensive look at the architecture of a genetically heterogeneous, phenotypically variable exemplar disorder, while at the same time, allowing the rational discovery of new therapeutic paradigms.

Public Health Relevance

In this proposal, we will dissect genetic interaction phenomena by studying the distribution of rare mutations across multiple Bardet-Biedl syndrome (BBS) genes and asking how such phenomena contribute to the development of endophenotypes in humans. In parallel, through the discovery of protective loci that have highlighted a role for the proteasome in the pathomechanism of BBS, we will perform a small molecule screen for the discovery of new candidate therapeutic leads. These studies will inform the genetic architecture of a disorder known for its genetic heterogeneity and clinical variability, while at the same time offering a rational route to the development of new therapies.

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Research Project (R01)
Project #
7R01HD042601-18
Application #
10090024
Study Section
Genetics of Health and Disease Study Section (GHD)
Program Officer
Parisi, Melissa
Project Start
2003-06-05
Project End
2024-03-31
Budget Start
2020-04-01
Budget End
2021-03-31
Support Year
18
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Children's Memorial Hospital (Chicago)
Department
Type
DUNS #
074438755
City
Chicago
State
IL
Country
United States
Zip Code
60611
Heydeck, Westley; Fievet, Lorraine; Davis, Erica E et al. (2018) The complexity of the cilium: spatiotemporal diversity of an ancient organelle. Curr Opin Cell Biol 55:139-149
Liu, Yangfan P; Bosch, Daniƫlle G M; Siemiatkowska, Anna M et al. (2017) Putative digenic inheritance of heterozygous RP1L1 and C2orf71 null mutations in syndromic retinal dystrophy. Ophthalmic Genet 38:127-132
Goetz, Sarah C; Bangs, Fiona; Barrington, Chloe L et al. (2017) The Meckel syndrome- associated protein MKS1 functionally interacts with components of the BBSome and IFT complexes to mediate ciliary trafficking and hedgehog signaling. PLoS One 12:e0173399
Helm, Benjamin M; Willer, Jason R; Sadeghpour, Azita et al. (2017) Partial uniparental isodisomy of chromosome 16 unmasks a deleterious biallelic mutation in IFT140 that causes Mainzer-Saldino syndrome. Hum Genomics 11:16
Frosk, Patrick; Arts, Heleen H; Philippe, Julien et al. (2017) A truncating mutation in CEP55 is the likely cause of MARCH, a novel syndrome affecting neuronal mitosis. J Med Genet 54:490-501
Ta-Shma, Asaf; Khan, Tahir N; Vivante, Asaf et al. (2017) Mutations in TMEM260 Cause a Pediatric Neurodevelopmental, Cardiac, and Renal Syndrome. Am J Hum Genet 100:666-675
Boldt, Karsten; van Reeuwijk, Jeroen; Lu, Qianhao et al. (2016) An organelle-specific protein landscape identifies novel diseases and molecular mechanisms. Nat Commun 7:11491
Katsanis, Nicholas (2016) The continuum of causality in human genetic disorders. Genome Biol 17:233
Lindstrand, Anna; Frangakis, Stephan; Carvalho, Claudia M B et al. (2016) Copy-Number Variation Contributes to the Mutational Load of Bardet-Biedl Syndrome. Am J Hum Genet 99:318-36
Oh, Edwin C; Vasanth, Shivakumar; Katsanis, Nicholas (2015) Metabolic regulation and energy homeostasis through the primary Cilium. Cell Metab 21:21-31

Showing the most recent 10 out of 65 publications