A saturation mutagenesis approach to understanding PTEN lipid phosphatase activity and genotype-phenotypes relationships

Phosphatase and tensin homolog (PTEN) is a tumor suppressor frequently mutated in diverse cancers. Germline PTEN mutations are also associated with a range of clinical outcomes, including PTEN hamartoma tumor syndrome (PHTS) and autism spectrum disorder (ASD). To empower new insights into PTEN function and clinically relevant genotype-phenotype relationships, we systematically evaluated the effect of PTEN mutations on lipid phosphatase activity in vivo. Using a massively parallel approach that leverages an artificial humanized yeast model, we derived high-confidence estimates of functional impact for 7,244 single amino acid PTEN variants (86% of possible). We identified 2,273 mutations with reduced cellular lipid phosphatase activity, which includes 1,789 missense mutations. These data recapitulated known functional findings but also uncovered new insights into PTEN protein structure, biochemistry, and mutation tolerance. Several residues in the catalytic pocket showed surprising mutational tolerance. We identified that the solvent exposure of wild-type residues is a critical determinant of mutational tolerance. Further, we created a comprehensive functional map by leveraging correlations between amino acid substitutions to impute functional scores for all variants, including those not present in the assay. Variant functional scores can reliably discriminate likely pathogenic from benign alleles. Further, 32% of ClinVar unclassified missense variants are phosphatase deficient in our assay, supporting their reclassification. ASD-associated mutations generally had less severe fitness scores relative to PHTS-associated mutations (p = 7.16 × 10-5) and a higher fraction of hypomorphic mutations, arguing for continued genotype-phenotype studies in larger clinical datasets that can further leverage these rich functional data.

[1]  Wonhwa Cho,et al.  Membrane-binding and activation mechanism of PTEN , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[2]  Víctor J. Cid,et al.  A Functional Dissection of PTEN N-Terminus: Implications in PTEN Subcellular Targeting and Tumor Suppressor Activity , 2015, PloS one.

[3]  David L. Young,et al.  Massively Parallel Functional Analysis of BRCA1 RING Domain Variants , 2015, Genetics.

[4]  R. Schiestl,et al.  High-efficiency yeast transformation using the LiAc/SS carrier DNA/PEG method , 2007, Nature Protocols.

[5]  Kali T. Witherspoon,et al.  Recurrent de novo mutations implicate novel genes underlying simplex autism risk , 2014, Nature Communications.

[6]  Atina G. Coté,et al.  A framework for exhaustively mapping functional missense variants , 2017, Molecular systems biology.

[7]  S. Kato,et al.  Understanding the function–structure and function–mutation relationships of p53 tumor suppressor protein by high-resolution missense mutation analysis , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[8]  C Eng,et al.  Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations , 2005, Journal of Medical Genetics.

[9]  Yuxin Yin,et al.  PTEN interacts with histone H1 and controls chromatin condensation. , 2014, Cell reports.

[10]  Pier Paolo Pandolfi,et al.  Subtle variations in Pten dose determine cancer susceptibility , 2010, Nature Genetics.

[11]  S. Fields,et al.  Deep mutational scanning: a new style of protein science , 2014, Nature Methods.

[12]  B. Luikart,et al.  The Role of PTEN in Neurodevelopment , 2020, Molecular Neuropsychiatry.

[13]  Marcel Martin Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .

[14]  Jay Shendure,et al.  Saturation Editing of Genomic Regions by Multiplex Homology-Directed Repair , 2014, Nature.

[15]  E. Braun,et al.  Phylogenetic Analyses of Sites in Different Protein Structural Environments Result in Distinct Placements of the Metazoan Root , 2020, Biology.

[16]  A. Andres-Pons,et al.  Assessment of PTEN tumor suppressor activity in nonmammalian models: the year of the yeast , 2008, Oncogene.

[17]  Francisca Vazquez,et al.  Phosphorylation of the PTEN Tail Regulates Protein Stability and Function , 2000, Molecular and Cellular Biology.

[18]  James Y. Zou Analysis of protein-coding genetic variation in 60,706 humans , 2015, Nature.

[20]  L. Amzel,et al.  Characterization of PTEN mutations in brain cancer reveals that pten mono-ubiquitination promotes protein stability and nuclear localization , 2017, Oncogene.

[21]  A. Knudson,et al.  A continuum model for tumour suppression , 2011, Nature.

[22]  Kara Dolinski,et al.  An extended set of yeast-based functional assays accurately identifies human disease mutations , 2016, Genome research.

[23]  A. Andres-Pons,et al.  Phosphatidylinositol 3-Kinase-dependent Activation of Mammalian Protein Kinase B/Akt in Saccharomyces cerevisiae, an in Vivo Model for the Functional Study of Akt Mutations* , 2009, Journal of Biological Chemistry.

[24]  Kali T. Witherspoon,et al.  Excess of rare, inherited truncating mutations in autism , 2015, Nature Genetics.

[25]  Heung-Chin Cheng,et al.  Unique biochemical properties of the protein tyrosine phosphatase activity of PTEN-demonstration of different active site structural requirements for phosphopeptide and phospholipid phosphatase activities of PTEN. , 2010, Biochimica et biophysica acta.

[26]  Maitreya J. Dunham,et al.  Variant Interpretation: Functional Assays to the Rescue. , 2017, American journal of human genetics.

[27]  K. McBride,et al.  The prevalence of PTEN mutations in a clinical pediatric cohort with autism spectrum disorders, developmental delay, and macrocephaly , 2009, Genetics in Medicine.

[28]  Erika L. Moen,et al.  Nuclear Excluded Autism-Associated Phosphatase and Tensin Homolog Mutations Dysregulate Neuronal Growth , 2017, Biological Psychiatry.

[29]  S. Henikoff,et al.  Amino acid substitution matrices from protein blocks. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[30]  Víctor J Cid,et al.  A comprehensive functional analysis of PTEN mutations: implications in tumor- and autism-related syndromes. , 2011, Human molecular genetics.

[31]  Jay Shendure,et al.  Quantitative Missense Variant Effect Prediction Using Large-Scale Mutagenesis Data. , 2017, Cell systems.

[32]  B. Frey,et al.  Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder , 2017, Nature Neuroscience.

[33]  Kandamurugu Manickam,et al.  Confirmation study of PTEN mutations among individuals with autism or developmental delays/mental retardation and macrocephaly , 2010, Autism research : official journal of the International Society for Autism Research.

[34]  W. Edelmann,et al.  SLiCE: a novel bacterial cell extract-based DNA cloning method , 2012, Nucleic acids research.

[35]  B. Eickholt,et al.  Functionally distinct groups of inherited PTEN mutations in autism and tumour syndromes , 2014, Journal of Medical Genetics.

[36]  Jing Li,et al.  Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome , 1997, Nature Genetics.

[37]  N. Leslie,et al.  Inherited PTEN mutations and the prediction of phenotype. , 2016, Seminars in cell & developmental biology.

[38]  T. Mikkelsen,et al.  Comprehensive mutational scanning of a kinase in vivo reveals substrate-dependent fitness landscapes , 2014, Nucleic acids research.

[39]  Ricardo Villamarín-Salomón,et al.  ClinVar: public archive of interpretations of clinically relevant variants , 2015, Nucleic Acids Res..

[40]  G. Bots,et al.  Lhermitte‐duclos disease and cowden disease: A single phakomatosis , 1991, Annals of neurology.

[41]  Bradley P. Coe,et al.  Multiplex Targeted Sequencing Identifies Recurrently Mutated Genes in Autism Spectrum Disorders , 2012, Science.

[42]  César Nombela,et al.  Reconstitution of the mammalian PI3K/PTEN/Akt pathway in yeast. , 2005, The Biochemical journal.

[43]  R. Pulido,et al.  Insights into the pathological mechanisms of p85α mutations using a yeast-based phosphatidylinositol 3-kinase model , 2017, Bioscience reports.

[44]  David L. Young,et al.  Deep mutational scanning of an RRM domain of the Saccharomyces cerevisiae poly(A)-binding protein , 2013, RNA.

[45]  C. Eng,et al.  Germline mutations in PTEN are present in Bannayan-Zonana syndrome , 1997, Nature Genetics.

[46]  Werner Braun,et al.  Exact and efficient analytical calculation of the accessible surface areas and their gradients for macromolecules , 1998 .

[47]  Anibal Gutierrez,et al.  SPARK: A US Cohort of 50,000 Families to Accelerate Autism Research , 2018, Neuron.

[48]  Inês Barroso,et al.  Prospective functional classification of all possible missense variants in PPARG , 2016, Nature Genetics.

[49]  S. Clarke,et al.  Deamidation, isomerization, and racemization at asparaginyl and aspartyl residues in peptides. Succinimide-linked reactions that contribute to protein degradation. , 1987, The Journal of biological chemistry.

[50]  Heung-Chin Cheng,et al.  PTEN catalysis of phospholipid dephosphorylation reaction follows a two-step mechanism in which the conserved aspartate-92 does not function as the general acid--mechanistic analysis of a familial Cowden disease-associated PTEN mutation. , 2007, Cellular signalling.

[51]  Sharmila Banerjee-Basu,et al.  SFARI Gene 2.0: a community-driven knowledgebase for the autism spectrum disorders (ASDs) , 2013, Molecular Autism.

[52]  Taylor L. Mighell,et al.  AAV-Mediated CRISPR/Cas9 Gene Editing in Murine Phenylketonuria , 2019, Molecular therapy. Methods & clinical development.

[53]  D. Baker,et al.  High Resolution Mapping of Protein Sequence–Function Relationships , 2010, Nature Methods.

[54]  Mingming Jia,et al.  COSMIC: somatic cancer genetics at high-resolution , 2016, Nucleic Acids Res..

[55]  Dmitrij Frishman,et al.  STRIDE: a web server for secondary structure assignment from known atomic coordinates of proteins , 2004, Nucleic Acids Res..

[56]  Moriah H Nissan,et al.  OncoKB: A Precision Oncology Knowledge Base. , 2017, JCO precision oncology.

[57]  B. Stec,et al.  Phospholipid-binding Sites of Phosphatase and Tensin Homolog (PTEN) , 2014, The Journal of Biological Chemistry.

[58]  Arne Gericke,et al.  A mutant form of PTEN linked to autism , 2010, Protein science : a publication of the Protein Society.

[59]  D. Cacchiarelli,et al.  Phenotypic Characterization of a Comprehensive Set of MAPK1/ERK2 Missense Mutants. , 2016, Cell reports.

[60]  Jiajie Zhang,et al.  PEAR: a fast and accurate Illumina Paired-End reAd mergeR , 2013, Bioinform..

[61]  C. Eng,et al.  Analysis of prevalence and degree of macrocephaly in patients with germline PTEN mutations and of brain weight in Pten knock-in murine model , 2011, European Journal of Human Genetics.

[62]  Terence P. Speed,et al.  Enrich2: a statistical framework for analyzing deep mutational scanning data , 2016, bioRxiv.

[63]  V. J. Cid,et al.  A Yeast-Based In Vivo Bioassay to Screen for Class I Phosphatidylinositol 3-Kinase Specific Inhibitors , 2012, Journal of biomolecular screening.

[64]  Vanessa E. Gray,et al.  Analysis of Large-Scale Mutagenesis Data To Assess the Impact of Single Amino Acid Substitutions , 2017, Genetics.

[65]  Itay Mayrose,et al.  ConSurf 2016: an improved methodology to estimate and visualize evolutionary conservation in macromolecules , 2016, Nucleic Acids Res..

[66]  Yuxin Yin,et al.  PTENβ is an alternatively translated isoform of PTEN that regulates rDNA transcription , 2017, Nature Communications.

[67]  W. Edelmann,et al.  Seamless Ligation Cloning Extract (SLiCE) cloning method. , 2014, Methods in molecular biology.

[68]  A. Andres-Pons,et al.  In vivo functional analysis of the counterbalance of hyperactive phosphatidylinositol 3-kinase p110 catalytic oncoproteins by the tumor suppressor PTEN. , 2007, Cancer research.

[69]  Laurence A. Turka,et al.  Cancer-Associated PTEN Mutants Act in a Dominant-Negative Manner to Suppress PTEN Protein Function , 2014, Cell.

[70]  C. Eng,et al.  Lifetime Cancer Risks in Individuals with Germline PTEN Mutations , 2012, Clinical Cancer Research.

[71]  J. Rubenstein,et al.  The parvalbumin/somatostatin ratio is increased in Pten mutant mice and by human PTEN ASD alleles. , 2015, Cell reports.

[72]  J. Shendure,et al.  Massively Parallel Genetics , 2016, Genetics.

[73]  Bale,et al.  Standards and Guidelines for the Interpretation of Sequence Variants: A Joint Consensus Recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology , 2015, Genetics in Medicine.

[74]  Tomohiko Maehama,et al.  Crystal Structure of the PTEN Tumor Suppressor Implications for Its Phosphoinositide Phosphatase Activity and Membrane Association , 1999, Cell.

[75]  R. B. Campbell,et al.  Allosteric Activation of PTEN Phosphatase by Phosphatidylinositol 4,5-Bisphosphate* , 2003, Journal of Biological Chemistry.

[76]  M. Wigler,et al.  The lipid phosphatase activity of PTEN is critical for its tumor supressor function. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[77]  J. Kitzman,et al.  Massively Parallel Single Amino Acid Mutagenesis , 2014, Nature Methods.

[78]  Ronald T Raines,et al.  Conformational stability and catalytic activity of PTEN variants linked to cancers and autism spectrum disorders. , 2015, Biochemistry.