80 results on '"Raymond T. O’Keefe"'
Search Results
2. Comparison of in silico strategies to prioritize rare genomic variants impacting RNA splicing for the diagnosis of genomic disorders
- Author
-
Charlie Rowlands, Huw B. Thomas, Jenny Lord, Htoo A. Wai, Gavin Arno, Glenda Beaman, Panagiotis Sergouniotis, Beatriz Gomes-Silva, Christopher Campbell, Nicole Gossan, Claire Hardcastle, Kevin Webb, Christopher O’Callaghan, Robert A. Hirst, Simon Ramsden, Elizabeth Jones, Jill Clayton-Smith, Andrew R. Webster, Genomics England Research Consortium, Andrew G. L. Douglas, Raymond T. O’Keefe, William G. Newman, Diana Baralle, Graeme C. M. Black, and Jamie M. Ellingford
- Subjects
Medicine ,Science - Abstract
Abstract The development of computational methods to assess pathogenicity of pre-messenger RNA splicing variants is critical for diagnosis of human disease. We assessed the capability of eight algorithms, and a consensus approach, to prioritize 249 variants of uncertain significance (VUSs) that underwent splicing functional analyses. The capability of algorithms to differentiate VUSs away from the immediate splice site as being ‘pathogenic’ or ‘benign’ is likely to have substantial impact on diagnostic testing. We show that SpliceAI is the best single strategy in this regard, but that combined usage of tools using a weighted approach can increase accuracy further. We incorporated prioritization strategies alongside diagnostic testing for rare disorders. We show that 15% of 2783 referred individuals carry rare variants expected to impact splicing that were not initially identified as ‘pathogenic’ or ‘likely pathogenic’; one in five of these cases could lead to new or refined diagnoses.
- Published
- 2021
- Full Text
- View/download PDF
3. Pathogenic Intronic Splice-Affecting Variants in MYBPC3 in Three Patients with Hypertrophic Cardiomyopathy
- Author
-
Katherine A. Wood, Jamie M. Ellingford, James Eden, Huw B. Thomas, Raymond T. O’Keefe, Claire Hopton, and William G. Newman
- Subjects
hypertrophic cardiomyopathy ,MYBPC3 ,splice variants ,minigene assays ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Genetic variants in MYBPC3 are one of the most common causes of hypertrophic cardiomyopathy (HCM). While variants in MYBPC3 affecting canonical splice site dinucleotides are a well-characterised cause of HCM, only recently has work begun to investigate the pathogenicity of more deeply intronic variants. Here, we present three patients with HCM and intronic splice-affecting MYBPC3 variants and analyse the impact of variants on splicing using in vitro minigene assays. We show that the three variants, a novel c.927-8G>A variant and the previously reported c.1624+4A>T and c.3815-10T>G variants, result in MYBPC3 splicing errors. Analysis of blood-derived patient RNA for the c.3815-10T>G variant revealed only wild type spliced product, indicating that mis-spliced transcripts from the mutant allele are degraded. These data indicate that the c.927-8G>A variant of uncertain significance and likely benign c.3815-10T>G should be reclassified as likely pathogenic. Furthermore, we find shortcomings in commonly applied bioinformatics strategies to prioritise variants impacting MYBPC3 splicing and re-emphasise the need for functional assessment of variants of uncertain significance in diagnostic testing.
- Published
- 2021
- Full Text
- View/download PDF
4. The Role of the U5 snRNP in Genetic Disorders and Cancer
- Author
-
Katherine A. Wood, Megan A. Eadsforth, William G. Newman, and Raymond T. O’Keefe
- Subjects
disease ,cancer ,U5 snRNP ,pre-mRNA splicing ,retinitis pigmentosa ,Burn-McKeown syndrome ,Genetics ,QH426-470 - Abstract
Pre-mRNA splicing is performed by the spliceosome, a dynamic macromolecular complex consisting of five small uridine-rich ribonucleoprotein complexes (the U1, U2, U4, U5, and U6 snRNPs) and numerous auxiliary splicing factors. A plethora of human disorders are caused by genetic variants affecting the function and/or expression of splicing factors, including the core snRNP proteins. Variants in the genes encoding proteins of the U5 snRNP cause two distinct and tissue-specific human disease phenotypes – variants in PRPF6, PRPF8, and SNRP200 are associated with retinitis pigmentosa (RP), while variants in EFTUD2 and TXNL4A cause the craniofacial disorders mandibulofacial dysostosis Guion-Almeida type (MFDGA) and Burn-McKeown syndrome (BMKS), respectively. Furthermore, recurrent somatic mutations or changes in the expression levels of a number of U5 snRNP proteins (PRPF6, PRPF8, EFTUD2, DDX23, and SNRNP40) have been associated with human cancers. How and why variants in ubiquitously expressed spliceosome proteins required for pre-mRNA splicing in all human cells result in tissue-restricted disease phenotypes is not clear. Additionally, why variants in different, yet interacting, proteins making up the same core spliceosome snRNP result in completely distinct disease outcomes – RP, craniofacial defects or cancer – is unclear. In this review, we define the roles of different U5 snRNP proteins in RP, craniofacial disorders and cancer, including how disease-associated genetic variants affect pre-mRNA splicing and the proposed disease mechanisms. We then propose potential hypotheses for how U5 snRNP variants cause tissue specificity resulting in the restricted and distinct human disorders.
- Published
- 2021
- Full Text
- View/download PDF
5. Assessment of disease-associated missense variants in RYR2 on transcript splicing
- Author
-
Damilola Olubando, Huw Thomas, Minoru Horie, Raymond T. O’Keefe, Luigi Venetucci, and William Newman
- Subjects
Genetic disease ,catecholaminergic ventricular tachycardia ,arrhythmia. ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Heterozygous RYR2 missense variants cause catecholaminergic polymorphic ventricular tachycardia. Rarely, loss of function variants can result in ventricular arrhythmias. We used splice prediction tools and an ex vivo splicing assay to investigate whether RYR2 missense variants result in altered splicing. Ten RYR2 variants were consistently predicted to disrupt splicing, however none altered splicing in the splicing assay. In summary, missense RYR2 variants are unlikely to cause disease by altered splicing.
- Published
- 2020
- Full Text
- View/download PDF
6. Bi-allelic FRA10AC1 variants in a multisystem human syndrome
- Author
-
Siddharth Banka, Stavit Shalev, Soo-Mi Park, Katherine A Wood, Huw B Thomas, Helen L Wright, Mohammed Alyahya, Sean Bankier, Ola Alimi, Elena Chervinsky, Leo A H Zeef, and Raymond T O’Keefe
- Subjects
Neurology (clinical) - Published
- 2022
- Full Text
- View/download PDF
7. 100,000 Genomes Pilot on Rare-Disease Diagnosis in Health Care — Preliminary Report
- Author
-
Gill Wilson, Anna de Burca, Marta Bleda, Lucy R. Wedderburn, Matthew Welland, Kathleen Stirrups, Valentina Cipriani, Kerrie Woods, Vijeya Ganesan, Susan Hill, Rosaline Quinlivan, Georgia Chan, Mehul T. Dattani, Robert McFarland, Graeme C.M. Black, Rutendo Mapeta, Augusto Rendon, Francesco Muntoni, James O.J. Davies, Mina Ryten, Rebecca E. Foulger, Arianna Tucci, Dina Halai, Tom Fowler, Noemi B.A. Roy, Sarah Leigh, Dragana Josifova, Philip Twiss, Ana L.T. Tavares, Zerin Hyder, Detlef Bockenhauer, Patrick Yu-Wai-Man, Lara Abulhoul, Nikolas Pontikos, Anthony T. Moore, Huw R. Morris, Patrick F. Chinnery, Nicholas W. Wood, Ellen A. Thomas, Shehla Mohammed, Sofia Douzgou, Tanya Lam, Kate Gibson, Robert Sarkany, Teofila Bueser, Wei Wei, Siddharth Banka, Alexander Broomfield, Hiva Fassihi, Nils Koelling, Carolyn Campbell, James Buchanan, Melita Irving, Sandrine Compeyrot-Lacassagne, Karola Rehmström, Austen Worth, Nikhil Thapar, Andrew R. Webster, Paul Brennan, Rita Horvath, Gavin Arno, Richard H Scott, Sam Malka, Andrew O.M. Wilkie, Sofie Ashford, Maria Bitner-Glindzicz, Jana Vandrovcova, William G. Newman, Caroline F. Wright, Andrew M. Schaefer, Roger F.L. James, Robert W. Taylor, Melanie Babcock, Arjune Sen, Emma Baple, Ellen M. McDonagh, Stephanie Grunewald, Loukas Moutsianas, Melissa A. Haendel, Olivera Spasic-Boskovic, Eleanor G. Seaby, Anna Need, Clarissa Pilkington, Sarah Wordsworth, Shamima Rahman, Christine Patch, Colin Wallis, Kristina Ibanez, Bishoy Habib, Eik Haraldsdottir, Huw B. Thomas, Razvan Sultana, Andrea H. Németh, Agata Wolejko, Claire Palles, Phil Beales, Adam C. Shaw, Letizia Vestito, Emily Li, Sarah Rose, Sarah Hunter, Angela Matchan, Genevieve Say, Dalia Kasperaviciute, Henry Houlden, Raymond T. O’Keefe, R. Andres Floto, Jill Clayton-Smith, John B. Taylor, Hywel J. Williams, Volker Straub, Val Davison, Helen Savage, John Chisholm, Eleanor Dewhurst, Charles Crichton, Andrea Haworth, Clare Turnbull, Carolyn Tregidgo, Carme Camps, Christopher Penkett, Emer O’Connor, Georgina Hall, Lyn S. Chitty, Sally Halsall, Andrew D. Mumford, Annette G. Wagner, Eleanor Williams, Mark Bale, Julius O. Jacobsen, Willem H. Ouwehand, Charu Deshpande, Gavin Burns, Smita Y. Patel, James Polke, Thiloka Ratnaike, Gavin Fuller, John Burn, Kenneth E. S. Poole, Emma Footitt, John R. Bradley, Suzanne Wood, Russell J. Grocock, Jenny C. Taylor, Louise Izatt, Kikkeri N. Naresh, Katherine R. Smith, Nigel Burrows, Katrina Newland, Peter N. Robinson, Sarju G. Mehta, Michael A. Simpson, Michael R. Barnes, Pilar Cacheiro, Olivia Niblock, Tracy Lester, Dimitris Polychronopoulos, Helen Brittain, John A. Sayer, Antonio Martin, Eshika Haque, Sean Humphray, Douglass M. Turnbull, Damian Smedley, Andrew Devereau, Stefan Gräf, Sian Ellard, Ivone U.S. Leong, Martin G. Reese, Matthias Wielscher, Louise C. Daugherty, Perry M. Elliott, F. Lucy Raymond, Cecilia Compton, David Bentley, Catherine Snow, James Welch, Frances Flinter, Dom McMullan, Mark J. Caulfield, Paul Aurora, Mark Gurnell, Mary Kasanicki, I. Karen Temple, Michel Michaelides, Deborah Ruddy, Leema Robert, Janice Yip, Grainne S. Gorman, Andrew C. Browning, Richard Quinton, Maureen Cleary, Jamie M. Ellingford, Angela Douglas, Christopher Boustred, and Investigators, The 100,000 Genomes Project Pilot
- Subjects
Adult ,Male ,Proband ,medicine.medical_specialty ,Adolescent ,Pilot Projects ,Genomics ,Polymerase Chain Reaction ,Genome ,State Medicine ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Health care ,Human Phenotype Ontology ,Humans ,Medicine ,Child ,Exome sequencing ,030304 developmental biology ,Family Characteristics ,0303 health sciences ,Whole Genome Sequencing ,Genome, Human ,business.industry ,Genetic Variation ,Rare Diseases/diagnosis ,General Medicine ,Middle Aged ,United Kingdom ,3. Good health ,Child, Preschool ,Family medicine ,Medical genetics ,Female ,business ,Bristol ,030217 neurology & neurosurgery ,Rare disease - Abstract
BACKGROUND: The U.K. 100,000 Genomes Project is in the process of investigating the role of genome sequencing in patients with undiagnosed rare diseases after usual care and the alignment of this research with health care implementation in the U.K. National Health Service. Other parts of this project focus on patients with cancer and infection.METHODS: We conducted a pilot study involving 4660 participants from 2183 families, among whom 161 disorders covering a broad spectrum of rare diseases were present. We collected data on clinical features with the use of Human Phenotype Ontology terms, undertook genome sequencing, applied automated variant prioritization on the basis of applied virtual gene panels and phenotypes, and identified novel pathogenic variants through research analysis.RESULTS: Diagnostic yields varied among family structures and were highest in family trios (both parents and a proband) and families with larger pedigrees. Diagnostic yields were much higher for disorders likely to have a monogenic cause (35%) than for disorders likely to have a complex cause (11%). Diagnostic yields for intellectual disability, hearing disorders, and vision disorders ranged from 40 to 55%. We made genetic diagnoses in 25% of the probands. A total of 14% of the diagnoses were made by means of the combination of research and automated approaches, which was critical for cases in which we found etiologic noncoding, structural, and mitochondrial genome variants and coding variants poorly covered by exome sequencing. Cohortwide burden testing across 57,000 genomes enabled the discovery of three new disease genes and 19 new associations. Of the genetic diagnoses that we made, 25% had immediate ramifications for clinical decision making for the patients or their relatives.CONCLUSIONS: Our pilot study of genome sequencing in a national health care system showed an increase in diagnostic yield across a range of rare diseases. (Funded by the National Institute for Health Research and others.).
- Published
- 2021
- Full Text
- View/download PDF
8. Eisosome disruption by noncoding RNA deletion increases protein secretion in yeast
- Author
-
Matthew Wenjie Feng, Daniela Delneri, Catherine B Millar, and Raymond T O'Keefe
- Abstract
Noncoding RNAs (ncRNAs) regulate many aspects of gene expression. We investigated how ncRNAs affected protein secretion in yeast by large-scale screening for improved endogenous invertase secretion in ncRNA deletion strains with deletion of stable unannotated transcripts (SUTs), cryptic unstable transcripts (CUTs), tRNAs, or snRNAs. We identified three candidate ncRNAs, SUT418, SUT390, and SUT125, that improved endogenous invertase secretion when deleted. As SUTs can affect expression of nearby genes, we quantified adjacent gene transcription and found that the PIL1 gene was down-regulated in the SUT125 deletion strain. Pil1 is a core component of eisosomes, nonmobile invaginations found throughout the plasma membrane. PIL1 knockout alone, or in combination with eisosome components LSP1 or SUR7, resulted in further increased secretion of invertase. Secretion of heterologous GFP was also increased upon PIL1 deletion, but this increase was signal sequence dependent. To reveal the potential for increased biopharmaceutical production, secretion of monoclonal antibody Pexelizumab scFv peptide was increased by PIL1 deletion. Global analysis of secreted proteins revealed that approximately 20% of secreted proteins, especially serine-enriched secreted proteins, including invertase, were increased upon eisosome disruption. Eisosomes are enriched with APC transporters and sphingolipids, which are essential components for secretory vesicle formation and protein sorting. Sphingolipid and serine biosynthesis pathways were up-regulated upon PIL1 deletion. We propose that increased secretion of endogenous and heterologous proteins upon PIL1 deletion resulted from sphingolipid redistribution in the plasma membrane and up-regulated sphingolipid biosynthesis. Overall, a new pathway to improve protein secretion in yeast via eisosome disruption has been identified.
- Published
- 2022
- Full Text
- View/download PDF
9. A basement membrane discovery pipeline uncovers network complexity, regulators, and human disease associations
- Author
-
Ranjay, Jayadev, Mychel R P T, Morais, Jamie M, Ellingford, Sandhya, Srinivasan, Richard W, Naylor, Craig, Lawless, Anna S, Li, Jack F, Ingham, Eric, Hastie, Qiuyi, Chi, Maryline, Fresquet, Nikki-Maria, Koudis, Huw B, Thomas, Raymond T, O'Keefe, Emily, Williams, Antony, Adamson, Helen M, Stuart, Siddharth, Banka, Damian, Smedley, David R, Sherwood, and Rachel, Lennon
- Subjects
Extracellular Matrix Proteins ,Multidisciplinary ,Basement Membrane/metabolism ,Extracellular Matrix/genetics ,Animals ,Humans ,Caenorhabditis elegans ,Caenorhabditis elegans/genetics ,Basement Membrane ,Zebrafish ,Extracellular Matrix Proteins/metabolism ,Zebrafish/genetics ,Extracellular Matrix - Abstract
Basement membranes (BMs) are ubiquitous extracellular matrices whose composition remains elusive, limiting our understanding of BM regulation and function. By developing a bioinformatic and in vivo discovery pipeline, we define a network of 222 human proteins and their animal orthologs localized to BMs. Network analysis and screening in C. elegans and zebrafish uncovered BM regulators, including ADAMTS, ROBO, and TGFβ. More than 100 BM network genes associate with human phenotypes, and by screening 63,039 genomes from families with rare disorders, we found loss-of-function variants in LAMA5 , MPZL2 , and MATN2 and show that they regulate BM composition and function. This cross-disciplinary study establishes the immense complexity of BMs and their impact on in human health.
- Published
- 2022
- Full Text
- View/download PDF
10. EFTUD2 missense variants disrupt protein function and splicing in mandibulofacial dysostosis Guion‐Almeida type
- Author
-
Huw B. Thomas, Kathryn E. Hentges, Christopher T. Gordon, William G. Newman, Vinod Varghese, Katherine A. Wood, Weronika A. Buczek, Jeanne Amiel, Raymond T. O'Keefe, Tania Attié-Bitach, and Veronique Pingault
- Subjects
Spliceosome ,Mandibulofacial Dysostosis Guion-Almeida type ,RNA Splicing ,Mutation, Missense ,Haploinsufficiency ,yeast ,Biology ,EFTUD2 ,03 medical and health sciences ,Pre-mRNA splicing ,Intellectual Disability ,Genetics ,Humans ,Missense mutation ,minigene ,Gene ,Ribonucleoprotein, U5 Small Nuclear ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,030305 genetics & heredity ,splicing variants ,Computational Biology ,Exons ,Peptide Elongation Factors ,Exon skipping ,missense variants ,RNA splicing ,Microcephaly ,Spliceosomes ,Snu114 ,Mandibulofacial Dysostosis ,Small nuclear ribonucleoprotein ,Minigene - Abstract
Pathogenic variants in the core spliceosome U5 small nuclear ribonucleoprotein gene EFTUD2/SNU114 cause the craniofacial disorder mandibulofacial dysostosis Guion-Almeida type (MFDGA). MFDGA-associated variants in EFTUD2 comprise large deletions encompassing EFTUD2, intragenic deletions and single nucleotide truncating or missense variants. These variants are predicted to result in haploinsufficiency by loss-of-function of the variant allele. While the contribution of deletions within EFTUD2 to allele loss-of-function are self-evident, the mechanisms by which missense variants are disease-causing have not been characterized functionally. Combining bioinformatics software prediction, yeast functional growth assays, and a minigene (MG) splicing assay, we have characterized how MFDGA missense variants result in EFTUD2 loss-of-function. Only four of 19 assessed missense variants cause EFTUD2 loss-of-function through altered protein function when modeled in yeast. Of the remaining 15 missense variants, five altered the normal splicing pattern of EFTUD2 pre-messenger RNA predominantly through exon skipping or cryptic splice site activation, leading to the introduction of a premature termination codon. Comparison of bioinformatic predictors for each missense variant revealed a disparity amongst different software packages and, in many cases, an inability to correctly predict changes in splicing subsequently determined by MG interrogation. This study highlights the need for laboratory-based validation of bioinformatic predictions for EFTUD2 missense variants.
- Published
- 2020
- Full Text
- View/download PDF
11. Global mapping of RNA homodimers in living cells
- Author
-
Marta M. Gabryelska, Andrew P. Badrock, Jian You Lau, Raymond T. O'Keefe, Yanick J. Crow, and Grzegorz Kudla
- Subjects
Base Sequence ,SARS-CoV-2 ,Zika Virus Infection ,Genetics ,Animals ,COVID-19 ,RNA, Small Nucleolar ,RNA, Viral ,Zika Virus ,Zebrafish ,Genetics (clinical) - Abstract
RNA homodimerization is important for various physiological processes, including the assembly of membraneless organelles, RNA subcellular localization, and packaging of viral genomes. However, understanding RNA dimerization has been hampered by the lack of systematic in vivo detection methods. Here, we show that CLASH, PARIS, and other RNA proximity ligation methods detect RNA homodimers transcriptome-wide as “overlapping” chimeric reads that contain more than one copy of the same sequence. Analyzing published proximity ligation data sets, we show that RNA:RNA homodimers mediated by direct base-pairing are rare across the human transcriptome, but highly enriched in specific transcripts, including U8 snoRNA, U2 snRNA, and a subset of tRNAs. Mutations in the homodimerization domain of U8 snoRNA impede dimerization in vitro and disrupt zebrafish development in vivo, suggesting an evolutionarily conserved role of this domain. Analysis of virus-infected cells reveals homodimerization of SARS-CoV-2 and Zika genomes, mediated by specific palindromic sequences located within protein-coding regions of N gene in SARS-CoV-2 and NS2A gene in Zika. We speculate that regions of viral genomes involved in homodimerization may constitute effective targets for antiviral therapies.
- Published
- 2022
- Full Text
- View/download PDF
12. Homozygous missense variants in BMPR15 can result in primary ovarian insufficiency
- Author
-
Leigh A.M. Demain, Kay Metcalfe, Eline Boetje, Peter Clayton, Elizabeth A. Martindale, Gail Busby, Raymond T. O'Keefe, and William G. Newman
- Subjects
Reproductive Medicine ,Adolescent ,Homozygote ,Mutation, Missense ,Obstetrics and Gynecology ,Humans ,Female ,Primary Ovarian Insufficiency ,Bone Morphogenetic Protein 15 ,Developmental Biology - Abstract
Does a genetic condition underlie the diagnosis of primary ovarian insufficiency (POI) in a 13-year-old girl with primary amenorrhoea?A case report of a next-generation sequencing panel of 24 genes associated with syndromal and non-syndromal POI was conducted.A homozygous missense variant c.1076CT, p.(Pro359Leu) in BMP15 was identified.The biallelic variant c.1076CT, p.(Pro359Leu) in BMP15 is associated with primary ovarian failure.
- Published
- 2022
13. Genome-wide analysis of the non-coding RNA synthetic genetic network reveals extensive plasticity and unique phenotypes in yeast
- Author
-
Marcin G Fraczek, Soukaina Timouma, Laura Natalia Balarezo-Cisneros, Steven Parker, Kobchai Duangrattanalert, Matthew Wenjie Feng, Sam Griffiths-Jones, Raymond T O’Keefe, and Daniela Delneri
- Abstract
Recent studies identified non-coding RNAs (ncRNAs) with unknown function that are responsible for major fitness changes in yeast. To understand ncRNA interplay and aid their functional assignment, the synthetic genetic array (SGA) methodology was employed to create >15,000 double mutants and to score their epistasis in different environments. Unlike the protein network, ncRNAs mostly displayed positive epistasis in rich medium. Interestingly, the negative interactions significantly increased under stressors, showing environmental-dependent functions for ncRNAs. No correlation was found between the network of ncRNAs and that of their neighbouring genes, suggesting functional independence. The U3 paralogs, SNR17A and SNR17B, share the majority of genetic interactions in rich medium as expected. For example, SUT480 interacted with both paralogs and its function was linked to 18S rRNA processing. However, under stressors, a large number of unique epistatic interactions were observed, supporting the notion that SNR17A and SNR17B have diverged and sub-functionalised after genome duplication.
- Published
- 2022
- Full Text
- View/download PDF
14. A basement membrane discovery pipeline uncovers network complexity, new regulators, and human disease associations
- Author
-
Nikki-Maria Koudis, Damian Smedley, Mychel Rpt Morais, Jamie M Ellingford, Antony Adamson, David R. Sherwood, Maryline Fresquet, Anna S. Li, Sandhya Srinivasan, Eric Hastie, Ranjay Jayadev, Richard W. Naylor, Rachel Lennon, Emily Williams, Raymond T. O'Keefe, Jack F. Ingham, Helen M. Stuart, Craig Lawless, Huw B. Thomas, Qiuyi Chi, and Siddharth Banka
- Subjects
Basement membrane ,Network complexity ,medicine.anatomical_structure ,biology ,ADAMTS ,medicine ,Computational biology ,biology.organism_classification ,Gene ,Genome ,Phenotype ,Zebrafish ,Function (biology) - Abstract
SummaryBasement membranes (BMs) are ubiquitous extracellular matrices whose composition remains elusive, limiting our understanding of BM regulation and function. By developing a bioinformatic and in vivo discovery pipeline, we define a network of 222 human proteins localized to BMs. Network analysis and screening in C. elegans and zebrafish identified new BM regulators, including ADAMTS, ROBO, and TGFβ. More than 100 BM-network genes associate with human phenotypes and by screening 63,039 genomes from families with rare disorders, we discovered loss-of-function variants in LAMA5, MPZL2, and MATN2, and show they regulate BM composition and function. This cross-disciplinary study establishes the immense complexity and role of BMs in human health.
- Published
- 2021
- Full Text
- View/download PDF
15. Expanding the genotypic spectrum of TXNL4A variants in Burn-McKeown syndrome
- Author
-
Jamie M Ellingford, Huw B. Thomas, Raymond T. O'Keefe, Glenda M. Beaman, Sofia Douzgou, William G. Newman, Katrina Prescott, Katherine A. Wood, and Emma Hobson
- Subjects
Heart Defects, Congenital ,Spliceosome ,Genotype ,RNA Splicing ,Biology ,Deafness ,Choanal Atresia ,Exon ,splicing ,Rare Disease ,Genetics ,Humans ,Genetic Predisposition to Disease ,Promoter Regions, Genetic ,Transcription factor ,Genetics (clinical) ,Alleles ,Genetic Association Studies ,Ribonucleoprotein, U5 Small Nuclear ,Binding Sites ,burn mckeown syndrome ,TNXL4A ,Facies ,Promoter ,Pedigree ,DNA binding site ,Phenotype ,RNA splicing ,Mutation ,Female ,Trans-acting ,Minigene ,Protein Binding ,Transcription Factors - Abstract
The developmental disorder Burn-McKeown Syndrome (BMKS) is characterised by choanal atresia and specific craniofacial features. BMKS is caused by biallelic variants in the pre-messenger RNA splicing factor TXNL4A. Most patients have a loss-of-function variant in trans with a 34-base pair (bp) deletion (type 1 Δ34) in the promoter region. Here, we identified two patients with BMKS. One individual has a TXNL4A c.93_94delCC, p.His32Argfs*21 variant combined with a type 1 Δ34 promoter deletion. The other has an intronic TXNL4A splice site variant (c.258-3C>G) and a type 1 Δ34 promoter deletion. We show the c.258-3C>G variant, and a previously reported c.258-2A>G variant, cause skipping of the final exon of TXNL4A in a minigene splicing assay. Furthermore, we identify putative transcription factor binding sites within the 56bp of the TXNL4A promoter affected by the type 1 and type 2 Δ34 and use dual luciferase assays to identify a 22bp repeated motif essential for TXNL4A expression within this promoter region. We propose that additional variants affecting critical transcription factor binding nucleotides within the 22bp repeated motif could be relevant to BMKS etiology. Finally, our data emphasizes the need to analyse the non-coding sequence in individuals where a single likely pathogenic coding variant is identified in an autosomal recessive disorder consistent with the clinical presentation.
- Published
- 2021
- Full Text
- View/download PDF
16. A homozygous missense variant in CHRM3 associated with familial urinary bladder disease
- Author
-
Adrian S. Woolf, William G. Newman, Ali Aishah, Katherine A. Wood, Raymond T. O'Keefe, Jill E. Urquhart, Glenda M. Beaman, Sanjeev S. Bhaskar, Gabriella Galatà, Helen M. Stuart, Keng Wee Teik, James O'Sullivan, and Huw B. Thomas
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Urinary system ,Mutation, Missense ,030105 genetics & heredity ,Gastroenterology ,Frameshift mutation ,03 medical and health sciences ,Internal medicine ,Prune belly ,Muscarinic acetylcholine receptor ,distended bladder ,Genetics ,medicine ,Mydriasis ,Missense mutation ,Humans ,Family ,Genetics (clinical) ,Receptor, Muscarinic M3 ,Base Sequence ,business.industry ,Homozygote ,Malaysia ,Urinary Bladder Diseases ,Original Articles ,CHRM3 ,urinary bladder disease ,medicine.disease ,030104 developmental biology ,prune belly ,Bladder Disorder ,Original Article ,Female ,medicine.symptom ,business ,Urinary bladder disease - Abstract
CHRM3 codes for the M3 muscarinic acetylcholine receptor that is located on the surface of smooth muscle cells of the detrusor, the muscle that effects urinary voiding. Previously, we reported brothers in a family affected by a congenital prune belly‐like syndrome with mydriasis due to homozygous CHRM3 frameshift variants. In this study, we describe two sisters with bladders that failed to empty completely and pupils that failed to constrict fully in response to light, who are homozygous for the missense CHRM3 variant c.352G > A; p.(Gly118Arg). Samples were not available for genotyping from their brother, who had a history of multiple urinary tract infections and underwent surgical bladder draining in the first year of life. He died at the age of 6 years. This is the first independent report of biallelic variants in CHRM3 in a family with a rare serious bladder disorder associated with mydriasis and provides important evidence of this association.
- Published
- 2019
- Full Text
- View/download PDF
17. Biallelic loss of function variants in STAG3 result in primary ovarian insufficiency
- Author
-
William G. Newman, Glenda M. Beaman, James O'Sullivan, Emma K. Miles, Raymond T. O'Keefe, Gail Busby, Jonathan J. Edgerley, Leigh A M Demain, Cheryl Fitzgerald, and Eline Boetje
- Subjects
media_common.quotation_subject ,Nonsense ,Cell Cycle Proteins ,Primary Ovarian Insufficiency ,Bioinformatics ,Young Adult ,medicine ,Humans ,Amenorrhea ,Gene ,Loss function ,Genetic testing ,media_common ,medicine.diagnostic_test ,business.industry ,Puberty ,High-Throughput Nucleotide Sequencing ,Obstetrics and Gynecology ,Karyotype ,medicine.disease ,Pedigree ,Fragile X syndrome ,Reproductive Medicine ,Codon, Nonsense ,Karyotyping ,Primary Ovarian Failure ,Mutation ,Female ,business ,Gene Deletion ,Developmental Biology ,Rare disease - Abstract
Research question Does a genetic condition underlie the diagnosis of primary ovarian insufficiency (POI) in a 21-year-old woman with primary amenorrhoea? Design A karyotype and genetic testing for Fragile X syndrome was undertaken. A next-generation sequencing panel of 24 genes associated with syndromal and non-syndromal POI was conducted. Results A nonsense variant c.1336G>T, p.(Glu446Ter) and whole gene deletion in STAG3 were identified. Conclusions Biallelic loss of function variants in STAG3 are associated with primary ovarian failure type 8 and are a rare cause of POI.
- Published
- 2021
- Full Text
- View/download PDF
18. New insights into Perrault syndrome, a clinically and genetically heterogeneous disorder
- Author
-
Alessandro Rea, Shoujun Gu, Michael Hoa, Sheikh Riazuddin, Asma A. Khan, Rabia Faridi, Raymond T. O'Keefe, Zunaira Munir, Thomas B. Friedman, Cristina Fenollar-Ferrer, Sadaf Naz, and William G. Newman
- Subjects
Male ,medicine.medical_specialty ,Hearing loss ,Hearing Loss, Sensorineural ,Cell Cycle Proteins ,Biology ,Amino Acyl-tRNA Synthetases ,Molecular genetics ,Genetics ,medicine ,Humans ,Child ,Genetics (clinical) ,Genetic heterogeneity ,medicine.disease ,Phenotype ,Human genetics ,Gonadal Dysgenesis, 46,XX ,Pedigree ,Mutation ,Etiology ,Sensorineural hearing loss ,Female ,medicine.symptom ,Age of onset - Abstract
Hearing loss and impaired fertility are common human disorders each with multiple genetic causes. Sometimes deafness and impaired fertility, which are the hallmarks of Perrault syndrome, co-occur in a person. Perrault syndrome is inherited as an autosomal recessive disorder characterized by bilateral mild to severe childhood sensorineural hearing loss with variable age of onset in both sexes and ovarian dysfunction in females who have a 46, XX karyotype. Since the initial clinical description of Perrault syndrome 70 years ago, the phenotype of some subjects may additionally involve developmental delay, intellectual deficit and other neurological disabilities, which can vary in severity in part dependent upon the genetic variants and the gene involved. Here, we review the molecular genetics and clinical phenotype of Perrault syndrome and focus on supporting evidence for the eight genes (CLPP, ERAL1, GGPS1, HARS2, HSD17B4, LARS2, RMND1, TWNK) associated with Perrault syndrome. Variants of these eight genes only account for approximately half of the individuals with clinical features of Perrault syndrome where the molecular genetic base remains under investigation. Additional environmental etiologies and novel Perrault disease-associated genes remain to be identified to account for unresolved cases. We also report a new genetic variant of CLPP, computational structural insight about CLPP and single cell RNAseq data for eight reported Perrault syndrome genes suggesting a common cellular pathophysiology for this disorder. Some unanswered questions are raised to kindle future research about Perrault syndrome.
- Published
- 2021
19. A KRAS-responsive long non-coding RNA controls microRNA processing
- Author
-
Hui Sun Leong, Lei Shi, Stefano Cairo, Francesca Galuppini, Sudhakar Sahoo, Gianpiero Di Leva, D. Smith, Raymond T. O'Keefe, Laura Brulle-Soumare, Matteo Fassan, Robert Sellers, Kang Zeng, Michela Garofalo, Stefano Volinia, Dave Lee, Athanasios R. Paliouras, Tiziana Monteverde, and Peter Magee
- Subjects
0301 basic medicine ,Lung Neoplasms ,endocrine system diseases ,General Physics and Astronomy ,Kaplan-Meier Estimate ,Mice, SCID ,medicine.disease_cause ,Microprocessor complex ,0302 clinical medicine ,Mice, Inbred NOD ,Carcinoma, Non-Small-Cell Lung ,Mice, Knockout ,Multidisciplinary ,Manchester Cancer Research Centre ,R735 ,Long non-coding RNA ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,RNA, Long Noncoding ,Female ,KRAS ,Nucleophosmin ,Lung Neoplasms/genetics ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Proto-Oncogene Proteins p21(ras) ,Proto-Oncogene Proteins p21(ras)/genetics ,03 medical and health sciences ,Carcinoma, Non-Small-Cell Lung/genetics ,Cell Line, Tumor ,microRNA ,medicine ,Gene silencing ,Animals ,Humans ,RNA, Long Noncoding/genetics ,Lung cancer ,neoplasms ,Gene Expression Profiling ,ResearchInstitutes_Networks_Beacons/mcrc ,Cancer ,General Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,respiratory tract diseases ,MicroRNAs ,MicroRNAs/genetics ,030104 developmental biology ,Gene Ontology ,Xenograft Model Antitumor Assays/methods ,A549 Cells ,Cancer research ,Long non-coding RNAs ,Gene Expression Profiling/methods ,Carcinogenesis ,Non-small-cell lung cancer - Abstract
Wild-type KRAS (KRASWT) amplification has been shown to be a secondary means of KRAS activation in cancer and associated with poor survival. Nevertheless, the precise role of KRASWT overexpression in lung cancer progression is largely unexplored. Here, we identify and characterize a KRAS-responsive lncRNA, KIMAT1 (ENSG00000228709) and show that it correlates with KRAS levels both in cell lines and in lung cancer specimens. Mechanistically, KIMAT1 is a MYC target and drives lung tumorigenesis by promoting the processing of oncogenic microRNAs (miRNAs) through DHX9 and NPM1 stabilization while halting the biogenesis of miRNAs with tumor suppressor function via MYC-dependent silencing of p21, a component of the Microprocessor Complex. KIMAT1 knockdown suppresses not only KRAS expression but also KRAS downstream signaling, thereby arresting lung cancer growth in vitro and in vivo. Taken together, this study uncovers a role for KIMAT1 in maintaining a positive feedback loop that sustains KRAS signaling during lung cancer progression and provides a proof of principle that interfering with KIMAT1 could be a strategy to hamper KRAS-induced tumorigenesis., Wild-type KRAS amplification is known to induce KRAS activation in cancer leading to poor prognostic outcomes. Here the authors identify a KRAS-responsive lncRNA, KIMAT1 that maintains KRAS signalling in lung cancer, suggesting that its targeting may prevent KRAS-driven tumourigenesis.
- Published
- 2021
- Full Text
- View/download PDF
20. MRSD: a novel quantitative approach for assessing suitability of RNA-seq in the clinical investigation of mis-splicing in Mendelian disease
- Author
-
Nicola Whiffin, William G. Newman, Jamie M Ellingford, Gillian I. Rice, Charlie Rowlands, H N Hall, Douglas Agl., Black Gcm., Tracy A Briggs, Algy Taylor, Raymond T. O'Keefe, Simon J. Hubbard, and Diana Baralle
- Subjects
symbols.namesake ,RNA splicing ,Metric (mathematics) ,Gene expression ,Mendelian inheritance ,symbols ,RNA-Seq ,Context (language use) ,Computational biology ,Biology ,Gene ,Deep sequencing - Abstract
BackgroundRNA-sequencing of patient biosamples is a promising approach to delineate the impact of genomic variants on splicing, but variable gene expression between tissues complicates selection of appropriate tissues. Relative expression level is often used as a metric to predict RNA-sequencing utility. Here, we describe a gene- and tissue-specific metric to inform the feasibility of RNA-sequencing, overcoming some issues with using expression values alone.ResultsWe derive a novel metric, Minimum Required Sequencing Depth (MRSD), for all genes across three human biosamples (whole blood, lymphoblastoid cell lines (LCLs) and skeletal muscle). MRSD estimates the depth of sequencing required from RNA-sequencing to achieve user-specified sequencing coverage of a gene, transcript or group of genes of interest. MRSD predicts levels of splice junction coverage with high precision (90.1-98.2%) and overcomes transcript region-specific sequencing biases. Applying MRSD scoring to established disease gene panels shows that LCLs are the optimum source of RNA, of the three investigated biosamples, for 69.3% of gene panels. Our approach demonstrates that up to 59.4% of variants of uncertain significance in ClinVar predicted to impact splicing could be functionally assayed by RNA-sequencing in at least one of the investigated biosamples.ConclusionsWe demonstrate the power of MRSD as a metric to inform choice of appropriate biosamples for the functional assessment of splicing aberrations. We apply MRSD in the context of Mendelian genetic disorders and illustrate its benefits over expression-based approaches. We anticipate that the integration of MRSD into clinical pipelines will improve variant interpretation and, ultimately, diagnostic yield.
- Published
- 2021
- Full Text
- View/download PDF
21. The role of splicing factors in retinitis pigmentosa: links to cilia
- Author
-
Sudipto Roy, Dale W Maxwell, Raymond T. O'Keefe, and Kathryn E. Hentges
- Subjects
RNA Splicing ,Disease ,Biology ,Biochemistry ,Ciliopathies ,Retina ,03 medical and health sciences ,Retinitis pigmentosa ,medicine ,RNA Precursors ,Animals ,Humans ,Genetic Predisposition to Disease ,Cilia ,030304 developmental biology ,Genetics ,0303 health sciences ,Cilium ,Point mutation ,030305 genetics & heredity ,medicine.disease ,Phenotype ,medicine.anatomical_structure ,RNA splicing ,Mutation ,Spliceosomes ,sense organs ,RNA Splicing Factors ,Retinitis Pigmentosa - Abstract
Cilia are critical to numerous biological functions, both in development and everyday homeostatic processes. Diseases arising from genetic mutations that cause cilia dysfunction are termed ciliopathies. Several ubiquitously expressed splicing factors have been implicated in the condition Retinitis Pigmentosa (RP), a group of diseases characterised by the progressive degeneration of the retina. In many types of RP the disease affects the modified primary cilium of the photoreceptor cells and thus, these types of RP are considered ciliopathies. Here, we discuss sequence variants found within a number of these splicing factors, the resulting phenotypes, and the mechanisms underpinning disease pathology. Additionally, we discuss recent evidence investigating why RP patients with mutations in globally expressed splicing factors present with retina-specific phenotypes.
- Published
- 2021
22. Functional and transcriptional profiling of non-coding RNAs in yeast reveal context-dependent phenotypes and in trans effects on the protein regulatory network
- Author
-
Daniela Delneri, Soukaina Timouma, Marcin G. Fraczek, Raymond T. O'Keefe, Catherine B. Millar, Steven Parker, Ping Wang, and Laura Natalia Balarezo-Cisneros
- Subjects
Cancer Research ,RNA, Untranslated ,Transcription, Genetic ,Mutagenesis and Gene Deletion Techniques ,RNA, Untranslated/genetics ,Cellular homeostasis ,Gene Expression ,Yeast and Fungal Models ,QH426-470 ,Haploidy ,Biochemistry ,Saccharomyces cerevisiae/genetics ,Gene Expression Regulation, Fungal ,Gene Regulatory Networks ,Small nucleolar RNA ,Genetics (clinical) ,Regulation of gene expression ,0303 health sciences ,Organic Compounds ,030302 biochemistry & molecular biology ,Eukaryota ,Transcriptome/genetics ,Non-coding RNA ,Nucleic acids ,Mutant Strains ,Chemistry ,Phenotypes ,Phenotype ,Experimental Organism Systems ,Physical Sciences ,Saccharomyces cerevisiae Proteins/genetics ,Genome, Fungal ,Research Article ,Saccharomyces cerevisiae Proteins ,Transcription Factors/genetics ,Context (language use) ,Computational biology ,Saccharomyces cerevisiae ,Steroid biosynthesis ,Biology ,Research and Analysis Methods ,03 medical and health sciences ,Saccharomyces ,Model Organisms ,Manchester Institute of Biotechnology ,Genetics ,Gene Expression Regulation, Fungal/genetics ,Gene Regulation ,RRNA processing ,Molecular Biology Techniques ,Gene ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Biology and life sciences ,Ethanol ,Deletion Mutagenesis ,Organic Chemistry ,Chemical Compounds ,Organisms ,Fungi ,ResearchInstitutes_Networks_Beacons/manchester_institute_of_biotechnology ,Yeast ,Alcohols ,Mutation ,Animal Studies ,RNA ,Transcriptome ,Transcription Factors ,Gene Regulatory Networks/genetics - Abstract
Non-coding RNAs (ncRNAs), including the more recently identified Stable Unannotated Transcripts (SUTs) and Cryptic Unstable Transcripts (CUTs), are increasingly being shown to play pivotal roles in the transcriptional and post-transcriptional regulation of genes in eukaryotes. Here, we carried out a large-scale screening of ncRNAs in Saccharomyces cerevisiae, and provide evidence for SUT and CUT function. Phenotypic data on 372 ncRNA deletion strains in 23 different growth conditions were collected, identifying ncRNAs responsible for significant cellular fitness changes. Transcriptome profiles were assembled for 18 haploid ncRNA deletion mutants and 2 essential ncRNA heterozygous deletants. Guided by the resulting RNA-seq data we analysed the genome-wide dysregulation of protein coding genes and non-coding transcripts. Novel functional ncRNAs, SUT125, SUT126, SUT035 and SUT532 that act in trans by modulating transcription factors were identified. Furthermore, we described the impact of SUTs and CUTs in modulating coding gene expression in response to different environmental conditions, regulating important biological process such as respiration (SUT125, SUT126, SUT035, SUT432), steroid biosynthesis (CUT494, SUT053, SUT468) or rRNA processing (SUT075 and snR30). Overall, these data capture and integrate the regulatory and phenotypic network of ncRNAs and protein-coding genes, providing genome-wide evidence of the impact of ncRNAs on cellular homeostasis., Author summary A quarter of the yeast genome comprises non-coding RNA molecules (ncRNAs), which do not translate into proteins but are involved in the regulation of gene expression. ncRNAs can affect nearby genes by physically interfering with their transcription (cis mode of action), or they interact with DNA, proteins or other RNAs to regulate the expression of distant genes (trans mode of action). Examples of cis-acting ncRNAs have been broadly described, however, genome-wide studies to identify functional trans-acting ncRNAs involved in global gene regulation are still lacking. Here, we used a ncRNA yeast deletion collection to score ncRNA impact on cellular function in different environmental conditions. A group of 20 ncRNA deletion mutants with broad fitness diversity were selected to investigate the ncRNA effect on the protein and ncRNA expression network. We showed a high correlation between altered phenotypes and global transcriptional changes, in an environmental dependent manner. We confirmed the trans acting regulation of ncRNAs in the genome and their role in altering the expression of transcription factors. These findings support the notion of the involvement of ncRNAs in fine tuning cellular expression via regulation of transcription factors, as an advantageous RNA-mediated mechanism that can be fast and cost-effective for the cells.
- Published
- 2021
- Full Text
- View/download PDF
23. Leukoencephalopathy with calcifications and cysts: Genetic and phenotypic spectrum
- Author
-
Calvin Soh, Sophie Calvert, Ram L. Kumar, Isabelle Desguerre, Kevin Talbot, Evangeline Wassmer, Axel Panzer, Andrea Berger, Anna de Burca, Anu Jacob, Andrea Whitney, Andrew P. Badrock, Frances Gibbon, Shelley MacDonald, Rhys H. Thomas, Reza Maroofian, Heather Burnett, Elizabeth Jones, Thomas Blauwblomme, Francois V. Bolduc, Jamal Ghoumid, Mickaël Ferrand, Yanick J. Crow, Emma M. Jenkinson, Camilo Toro, Diana Chiang, Roseline Caumes, Gillian I. Rice, Gemma Fisher, Gopinath M. Subramanian, Edoardo Monfrini, Renaud Touraine, Hilde T. Hilmarsen, Sarju G. Mehta, Imelda Hughes, Sumit Parikh, Edward Blair, Mary O'Driscoll, Sarah Dyack, Himanshu Goel, Kristin W. Barañano, Prab Prabhakar, Luis Seabra, Roberta Battini, John H. Livingston, Russell P. Saneto, Richard J. Leventer, Katrin Õunap, Heather Marshall, Andy Cheuk Him Ng, Duccio Maria Cordelli, Natasha Demic, Daniela Neumann, Natalie Boddaert, Michael J. Noetzel, S. Richard Dunham, Ehsan Ghayoor Karimiani, Johannes A. Buckard, Frances Elmslie, Raymond T. O'Keefe, Chloe A Stutterd, Richard Sandford, Imke Metz, Francis Ramond, Liesbeth De Waele, Alessio Di Fonzo, Emma Wakeling, David B. Clifford, Crow Y.J., Marshall H., Rice G.I., Seabra L., Jenkinson E.M., Baranano K., Battini R., Berger A., Blair E., Blauwblomme T., Bolduc F., Boddaert N., Buckard J., Burnett H., Calvert S., Caumes R., Ng A.C.-H., Chiang D., Clifford D.B., Cordelli D.M., de Burca A., Demic N., Desguerre I., De Waele L., Di Fonzo A., Dunham S.R., Dyack S., Elmslie F., Ferrand M., Fisher G., Karimiani E.G., Ghoumid J., Gibbon F., Goel H., Hilmarsen H.T., Hughes I., Jacob A., Jones E.A., Kumar R., Leventer R.J., MacDonald S., Maroofian R., Mehta S.G., Metz I., Monfrini E., Neumann D., Noetzel M., O'Driscoll M., Ounap K., Panzer A., Parikh S., Prabhakar P., Ramond F., Sandford R., Saneto R., Soh C., Stutterd C.A., Subramanian G.M., Talbot K., Thomas R.H., Toro C., Touraine R., Wakeling E., Wassmer E., Whitney A., Livingston J.H., O'Keefe R.T., and Badrock A.P.
- Subjects
Male ,0301 basic medicine ,Proband ,030105 genetics & heredity ,Gene mutation ,ribosomopathy ,Compound heterozygosity ,Genetic analysis ,Loss of heterozygosity ,Leukoencephalopathy ,Consanguinity ,Leukoencephalopathies ,Pathology, Molecular ,Child ,Zebrafish ,Genetics (clinical) ,Genetics ,Molecular pathology ,C/D box snoRNA U8 ,coats plus ,Labrune syndrome ,leukoencephalopathy with calcifications and cysts ,SNORD118 ,Calcinosis ,Middle Aged ,3. Good health ,Child, Preschool ,Female ,Adult ,Heterozygote ,Adolescent ,coats plu ,Biology ,Young Adult ,03 medical and health sciences ,medicine ,Animals ,Humans ,RNA, Small Nucleolar ,Genetic Association Studies ,Aged ,leukoencephalopathy with calcifications and cyst ,Infant, Newborn ,Infant ,medicine.disease ,Disease Models, Animal ,030104 developmental biology - Abstract
Biallelic mutations in SNORD118, encoding the small nucleolar RNA U8, cause leukoencephalopathy with calcifications and cysts (LCC). Given the difficulty in interpreting the functional consequences of variants in nonprotein encoding genes, and the high allelic polymorphism across SNORD118 in controls, we set out to provide a description of the molecular pathology and clinical spectrum observed in a cohort of patients with LCC. We identified 64 affected individuals from 56 families. Age at presentation varied from 3 weeks to 67 years, with disease onset after age 40 years in eight patients. Ten patients had died. We recorded 44 distinct, likely pathogenic, variants in SNORD118. Fifty two of 56 probands were compound heterozygotes, with parental consanguinity reported in only three families. Forty nine of 56 probands were either heterozygous (46) or homozygous (three) for a mutation involving one of seven nucleotides that facilitate a novel intramolecular interaction between the 5' end and 3' extension of precursor-U8. There was no obvious genotype-phenotype correlation to explain the marked variability in age at onset. Complementing recently published functional analyses in a zebrafish model, these data suggest that LCC most often occurs due to combinatorial severe and milder mutations, with the latter mostly affecting 3' end processing of precursor-U8.
- Published
- 2021
24. Uncovering genetic mechanisms of hypertension through multi-omic analysis of the kidney
- Author
-
Ingrid Wise, Bradley Godfrey, Raymond T. O'Keefe, Mikael Ekholm, Wojciech Wystrychowski, Pasquale Maffia, Matthias Kretzler, Sushant Saluja, James Eales, Artur Akbarov, Christopher Finan, Maciej Tomaszewski, Monika Szulińska, Gosia Trynka, Matthew Denniff, Sanjeev Pramanik, Ewa Zukowska-Szczechowska, Bernard Keavney, Andrew P. Morris, Yusif Shakanti, Sandesh Chopade, John Bowes, Eddie Cano-Gamez, Huw B. Thomas, Matthew G. Sampson, Xiaoguang Xu, Evangelos Evangelou, Paweł Bogdański, Priscilla R. Prestes, Stephen Eyre, Xiao Jiang, David Talavera, Fadi J. Charchar, Hui Guo, Andrzej Antczak, Joanna Zywiec, Nilesh J. Samani, Alicja Nazgiewicz, Michelle T. McNulty, Adrian S. Woolf, Robert Król, Tomasz J. Guzik, Jason Brown, Carlo Berzuini, Mahan Salehi, Maciej Glyda, Aroon D. Hingorani, Felix Eichinger, Mark J. Caulfield, Eales, J. M., Jiang, X., Xu, X., Saluja, S., Akbarov, A., Cano-Gamez, E., Mcnulty, M. T., Finan, C., Guo, H., Wystrychowski, W., Szulinska, M., Thomas, H. B., Pramanik, S., Chopade, S., Prestes, P. R., Wise, I., Evangelou, E., Salehi, M., Shakanti, Y., Ekholm, M., Denniff, M., Nazgiewicz, A., Eichinger, F., Godfrey, B., Antczak, A., Glyda, M., Krol, R., Eyre, S., Brown, J., Berzuini, C., Bowes, J., Caulfield, M., Zukowska-Szczechowska, E., Zywiec, J., Bogdanski, P., Kretzler, M., Woolf, A. S., Talavera, D., Keavney, B., Maffia, P., Guzik, T. J., O'Keefe, R. T., Trynka, G., Samani, N. J., Hingorani, A., Sampson, M. G., Morris, A. P., Charchar, F. J., and Tomaszewski, M.
- Subjects
Quantitative Trait Loci ,Genome-wide association study ,Blood Pressure ,Quantitative trait locus ,Biology ,Kidney ,Polymorphism, Single Nucleotide ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Mendelian randomization ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Mendelian Randomization Analysi ,030304 developmental biology ,0303 health sciences ,Genetic Variation ,Mendelian Randomization Analysis ,Epigenome ,Genomics ,DNA Methylation ,Alternative Splicing ,medicine.anatomical_structure ,DNA methylation ,Hypertension ,030217 neurology & neurosurgery ,Human ,Genome-Wide Association Study - Abstract
The kidney is an organ of key relevance to blood pressure (BP) regulation, hypertension and antihypertensive treatment. However, genetically mediated renal mechanisms underlying susceptibility to hypertension remain poorly understood. We integrated genotype, gene expression, alternative splicing and DNA methylation profiles of up to 430 human kidneys to characterize the effects of BP index variants from genome-wide association studies (GWASs) on renal transcriptome and epigenome. We uncovered kidney targets for 479 (58.3%) BP-GWAS variants and paired 49 BP-GWAS kidney genes with 210 licensed drugs. Our colocalization and Mendelian randomization analyses identified 179 unique kidney genes with evidence of putatively causal effects on BP. Through Mendelian randomization, we also uncovered effects of BP on renal outcomes commonly affecting patients with hypertension. Collectively, our studies identified genetic variants, kidney genes, molecular mechanisms and biological pathways of key relevance to the genetic regulation of BP and inherited susceptibility to hypertension.
- Published
- 2021
- Full Text
- View/download PDF
25. Bi-allelic variants in the mitochondrial RNase P subunit PRORP cause mitochondrial tRNA processing defects and pleiotropic multisystem presentations
- Author
-
Brendan J. Battersby, Julie Richer, Leigh A M Demain, William G. Newman, Inna A. Belyantseva, Meredith K. Gillespie, Stephanie Oerum, Jill E. Urquhart, Thomas B. Friedman, James O'Sullivan, Alessandro Rea, Agatha Schlüter, Simon G. Williams, Hugh J. McMillan, Kevin J. Munro, Albert Amberger, Waheeda Pagarkar, Melanie Barzik, Kyle Thompson, Walter Rossmanith, Agustí Rodríguez-Palmero, Irit Hochberg, Sanjeev S. Bhaskar, Reeya Motha, Raymond T. O'Keefe, Zeev Blumenfeld, Pilar Quijada-Fraile, Edgard Verdura, Wyatt W. Yue, Johannes Zschocke, Sandra Demetz, Andrea J. Deutschmann, Aurora Pujol, Jessica L. Zambonin, Glenda M. Beaman, Isabella R. Lawrence, Kah Ying Ng, Sergey Yalonetsky, Emma M. Jenkinson, Robert W. Taylor, Institute of Biotechnology, and Biosciences
- Subjects
Male ,leukodystrophy ,RNA, Mitochondrial ,Ribonuclease P/genetics ,Mitochondrion ,sensorineural hearing loss ,RNase P ,0302 clinical medicine ,RNA, Transfer ,MRPP3 ,PRORP ,Mitochondrial tRNA processing ,Genetics (clinical) ,Genetics ,0303 health sciences ,1184 Genetics, developmental biology, physiology ,mitochondria ,Genetic Pleiotropy ,Phenotype ,Perrault syndrome ,Pedigree ,Rare diseases ,Mitochondria ,Sensorineural hearing loss ,Female ,Malalties rares ,RNA, Transfer/genetics ,Adult ,Mitochondrial RNA processing ,Protein subunit ,rare disease ,Primary ovarian insufficiency ,Biology ,Ovary diseases ,Ribonuclease P ,03 medical and health sciences ,Report ,Complementary DNA ,Humans ,Allele ,Alleles ,030304 developmental biology ,Malalties de l'ovari ,MUTATIONS ,primary ovarian insufficiency ,Leukodystrophy ,HSD10 DISEASE ,MODEL ,RNA, Mitochondrial/genetics ,Mitochondria/enzymology ,Rare disease ,030217 neurology & neurosurgery - Abstract
Human mitochondrial RNase P (mt-RNase P) is responsible for 5' end processing of mitochondrial precursor tRNAs, a vital step in mitochondrial RNA maturation, and is comprised of three protein subunits: TRMT10C, SDR5C1 (HSD10), and PRORP. Pathogenic variants in TRMT10C and SDR5C1 are associated with distinct recessive or x-linked infantile onset disorders, resulting from defects in mitochondrial RNA processing. We report four unrelated families with multisystem disease associated with bi-allelic variants in PRORP, the metallonuclease subunit of mt-RNase P. Affected individuals presented with variable phenotypes comprising sensorineural hearing loss, primary ovarian insufficiency, developmental delay, and brain white matter changes. Fibroblasts from affected individuals in two families demonstrated decreased steady state levels of PRORP, an accumulation of unprocessed mitochondrial transcripts, and decreased steady state levels of mitochondrial-encoded proteins, which were rescued by introduction of the wild-type PRORP cDNA. In mt-tRNA processing assays performed with recombinant mt-RNase P proteins, the disease-associated variants resulted in diminished mitochondrial tRNA processing. Identification of disease-causing variants in PRORP indicates that pathogenic variants in all three subunits of mt-RNase P can cause mitochondrial dysfunction, each with distinct pleiotropic clinical presentations.
- Published
- 2021
- Full Text
- View/download PDF
26. Comparison of in silico strategies to prioritize rare genomic variants impacting RNA splicing for the diagnosis of genomic disorders
- Author
-
Augusto Rendon, A. Kousathanas, S. E. A. Leigh, D. Kasperaviciute, R. Jackson, J. Pullinger, Richard H. Scott, T. Rahim, Graeme C.M. Black, C. A. Odhams, Simon C Ramsden, A. Siddiq, L. Lahnstein, S. R. Thompson, Huw B. Thomas, Jenny Lord, M. Bleda, M. J. Welland, L. Moutsianas, A. Giess, Jill Clayton-Smith, A. Stuckey, Panagiotis I. Sergouniotis, H. Brittain, K. Sawant, Arianna Tucci, A. Sosinsky, I. U. S. Leong, Nicole Gossan, William G. Newman, Christopher Campbell, Mark J. Caulfield, T. Rogers, Diana Baralle, Andrew G. L. Douglas, A. L. Taylor Tavares, N. Murugaesu, Gavin Arno, S. M. Wood, Louise J. Jones, Robert A. Hirst, Htoo A Wai, A. Hamblin, Glenda M. Beaman, P. O’Donovan, A. Sieghart, F. Maleady-Crowe, S. Henderson, M. Tanguy, Claire Hardcastle, C. R. Boustred, G. C. Chan, M. McEntagart, Beatriz Gomes-Silva, E. Williams, Andrew R. Webster, Ellen R A Thomas, T. Fowler, Christine Patch, Raymond T. O'Keefe, Elizabeth A. Jones, D. Perez-Gil, M. B. Pereira, R. Bevers, F. J. Lopez, Jamie M Ellingford, Kevin Webb, M. Kayikci, S. C. Smith, F. Boardman-Pretty, Charlie Rowlands, K. Witkowsa, P. Arumugam, A. C. Need, Tim Hubbard, J. C. Ambrose, M. Mueller, Christopher O'Callaghan, F. Minneci, and K. Savage
- Subjects
Prioritization ,Genetic testing ,Science ,RNA Splicing ,In silico ,Computational biology ,Biology ,Article ,Diagnosis, Differential ,Human disease ,Databases, Genetic ,Diagnosis ,RNA Precursors ,Genetics ,Humans ,Disease ,Clinical genetics ,Medical diagnosis ,Uncertain significance ,Diagnostic Techniques and Procedures ,Multidisciplinary ,Disease genetics ,Computational Biology ,Genetic Variation ,Diagnostic test ,Exons ,Genomics ,Pathogenicity ,Mutation ,RNA splicing ,Medicine ,RNA Splice Sites ,Algorithms - Abstract
The development of computational methods to assess pathogenicity of pre-messenger RNA splicing variants is critical for diagnosis of human disease. We assessed the capability of eight algorithms, and a consensus approach, to prioritize 250 variants of uncertain significance (VUSs) that underwent splicing functional analyses. It is the capability of algorithms to differentiate VUSs away from the immediate splice site as being ‘pathogenic’ or ‘benign’ that is likely to have the most substantial impact on diagnostic testing. We show that SpliceAI is the best single strategy in this regard, but that combined usage of tools using a weighted approach can increase accuracy further. We incorporated prioritization strategies alongside diagnostic testing for rare disorders. We show that 15% of 2783 referred individuals carry rare variants expected to impact splicing that were not initially identified as ‘pathogenic’ or ‘likely pathogenic’; 1 in 5 of these cases could lead to new or refined diagnoses.
- Published
- 2020
- Full Text
- View/download PDF
27. Functional and transcriptional profiling of non-coding RNAs in yeast reveal context-dependent phenotypes and widespread in trans effects on the protein regulatory network
- Author
-
Ping Wang, Laura Natalia Balarezo-Cisneros, Soukaina Timouma, Catherine B. Millar, Marcin G. Fraczek, Daniela Delneri, Steven Parker, and Raymond T. O'Keefe
- Subjects
Regulation of gene expression ,Gene expression ,Cellular homeostasis ,Computational biology ,Steroid biosynthesis ,Biology ,RRNA processing ,Non-coding RNA ,Transcription factor ,Gene - Abstract
Non-coding RNAs (ncRNAs), including the more recently identified Stable Unannotated Transcripts (SUTs) and Cryptic Unstable Transcripts (CUTs), are increasingly being shown to play pivotal roles in the transcriptional and post-transcriptional regulation of genes in eukaryotes. Here, we carried out a large-scale screening of ncRNAs in Saccharomyces cerevisiae, and provide evidence for SUT and CUT function. Phenotypic data on 372 ncRNA deletion strains in 23 different growth conditions were collected, identifying ncRNAs responsible for significant cellular fitness changes. Transcriptome profiles were assembled for 18 haploid ncRNA deletion mutants and 2 essential ncRNA heterozygous deletants. Guided by the resulting RNA-seq data we analysed the genome-wide dysregulation of protein coding genes and non-coding transcripts. Novel functional ncRNAs, SUT125, SUT126, SUT035 and SUT532 that act in trans by modulating transcription factors were identified. Furthermore, we described the impact of SUTs and CUTs in modulating coding gene expression in response of different environmental conditions, regulating important biological process such as respiration (SUT125, SUT126, SUT035, SUT432), steroid biosynthesis (CUT494, SUT530, SUT468) or rRNA processing (SUT075 and snR30). Overall, this data captures and integrates the regulatory and phenotypic network of ncRNAs and protein coding genes, providing genome-wide evidence of the impact of ncRNAs on cellular homeostasis.Author SummaryThe yeast genome contains 25% of non-coding RNA molecules (ncRNAs), which do not translate into proteins but are involved in regulation of gene expression. ncRNAs can affect nearby genes by physically interfering with their transcription (cis mode of action), or they interact with DNA, proteins or others RNAs to regulate the expression of distant genes (trans mode of action). Examples of cis-acting ncRNAs have been broadly described, however genome-wide studies to identify functional trans-acting ncRNAs involved in global gene regulation are still lacking. Here, we used the ncRNA yeast deletion collection to score their impact on cellular function in different environmental conditions. A group of 20 ncRNAs mutants with broad fitness diversity were selected to investigate their effect on the protein and ncRNA expression network. We showed a high correlation between altered phenotypes and global transcriptional changes, in an environmental dependent manner. We confirmed the widespread trans acting expressional regulation of ncRNAs in the genome and their role in affecting transcription factors. These findings support the notion of the involvement on ncRNAs in fine tuning the cellular expression via regulations of TFs, as an advantageous RNA-mediated mechanism that can be fast and cost-effective for the cells.
- Published
- 2020
- Full Text
- View/download PDF
28. Analysis of U8 snoRNA Variants in Zebrafish Reveals How Bi-allelic Variants Cause Leukoencephalopathy with Calcifications and Cysts
- Author
-
Emma M. Jenkinson, Andrew P. Badrock, Denis L. J. Lafontaine, Gillian I. Rice, Raymond T. O'Keefe, Siobhan Crilly, Paul R. Kasher, Ludivine Wacheul, Carolina Uggenti, and Yanick J. Crow
- Subjects
Génétique clinique ,Ribosomopathy ,Mutant ,Ribosome biogenesis ,ribosomopathy ,Conserved sequence ,U8 snoRNA ,0302 clinical medicine ,Leukoencephalopathies ,Embryonic Development/genetics ,Central Nervous System Cysts/genetics ,Central Nervous System Cysts ,Zebrafish ,Genetics (clinical) ,Conserved Sequence ,Genetics ,0303 health sciences ,Leukoencephalopathies/genetics ,Cysts ,Calcinosis ,Tumor Suppressor Protein p53/genetics ,Labrune syndrome ,Haploinsufficiency ,Biologie ,leukoencephalopathy with calcifications and cysts ,Embryonic Development ,Biology ,snoRNA ,Article ,SNORD118 ,03 medical and health sciences ,Zebrafish Proteins/genetics ,Animals ,Humans ,RNA, Small Nucleolar ,Allele ,RRNA processing ,Alleles ,030304 developmental biology ,Base Sequence ,Calcinosis/genetics ,Cysts/genetics ,Zebrafish Proteins ,zebrafish ,biology.organism_classification ,Disease Models, Animal ,Mutation ,Zebrafish/embryology ,Tumor Suppressor Protein p53 ,030217 neurology & neurosurgery ,RNA, Small Nucleolar/genetics - Abstract
How mutations in the non-coding U8 snoRNA cause the neurological disorder leukoencephalopathy with calcifications and cysts (LCC) is poorly understood. Here, we report the generation of a mutant U8 animal model for interrogating LCC-associated pathology. Mutant U8 zebrafish exhibit defective central nervous system development, a disturbance of ribosomal RNA (rRNA) biogenesis and tp53 activation, which monitors ribosome biogenesis. Further, we demonstrate that fibroblasts from individuals with LCC are defective in rRNA processing. Human precursor-U8 (pre-U8) containing a 3′ extension rescued mutant U8 zebrafish, and this result indicates conserved biological function. Analysis of LCC-associated U8 mutations in zebrafish revealed that one null and one functional allele contribute to LCC. We show that mutations in three nucleotides at the 5′ end of pre-U8 alter the processing of the 3′ extension, and we identify a previously unknown base-pairing interaction between the 5′ end and the 3′ extension of human pre-U8. Indeed, LCC-associated mutations in any one of seven nucleotides in the 5′ end and 3′ extension alter the processing of pre-U8, and these mutations are present on a single allele in almost all individuals with LCC identified to date. Given genetic data indicating that bi-allelic null U8 alleles are likely incompatible with human development, and that LCC is not caused by haploinsufficiency, the identification of hypomorphic misprocessing mutations that mediate viable embryogenesis furthers our understanding of LCC molecular pathology and cerebral vascular homeostasis., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2020
- Full Text
- View/download PDF
29. Novel intramolecular base-pairing of the U8 snoRNA underlies a Mendelian form of cerebral small vessel disease
- Author
-
Siobhan Crilly, Denis L. J. Lafontaine, Ludivine Wacheul, Gillian I. Rice, Emma M. Jenkinson, Raymond T. O'Keefe, Yanick J. Crow, Paul R. Kasher, Carolina Uggenti, and Andrew P. Badrock
- Subjects
Genetics ,biology ,Mutant ,Ribosome biogenesis ,Ribosomal RNA ,Small nucleolar RNA ,Allele ,Sciences bio-médicales et agricoles ,RRNA processing ,biology.organism_classification ,Zebrafish ,Biogenesis - Abstract
How mutations in the non-coding U8 snoRNA cause the neurological disorder leukoencephalopathy with calcification and cysts (LCC) is poorly understood. We report the first vertebrate mutant U8 animal model for interrogating LCC-associated pathology. Mutant U8 zebrafish exhibit defective central nervous system development and ribosomal RNA (rRNA) biogenesis, with tp53 activation which monitors ribosome biogenesis. Importantly, LCC patient fibroblasts demonstrate rRNA processing defects. Human precursor-U8 (pre-U8) containing a 3’ extension rescued mutant U8 zebrafish, indicating conserved biological function. Analysis of LCC-associated U8 alleles in zebrafish revealed that one null and one hypomorphic, but still functional, allele combine to cause LCC. Mutations involving any one of seven nucleotides within the human pre-U8 3’ extension, or 5’ region of U8, alter processing of pre-U8, and identify a novel base-pairing interaction between the 5’ end and 3’ extension of human pre-U8. Variants in these seven nucleotides, one of which is present on a single allele in almost all patients, act as hypomorphic mutations. Given that biallelic null U8 alleles are likely incompatible with human development, identification of hypomorphic mutations mediating viable embryogenesis furthers understanding of LCC molecular pathology and cerebral vascular homeostasis.
- Published
- 2019
- Full Text
- View/download PDF
30. Advanced Methods for the Analysis of Altered Pre-mRNA Splicing in Yeast and Disease
- Author
-
Huw B, Thomas and Raymond T, O'Keefe
- Subjects
RNA Splicing ,RNA Precursors ,Spliceosomes ,Animals ,Humans ,Saccharomyces cerevisiae - Abstract
Splicing of pre-messenger RNA (pre-mRNA) transcripts is a fundamental process in all eukaryotes that provides a mechanism of increasing the proteomic diversity within a cell that can be tightly regulated in a dynamic manner. While constitutive and alternative splicing are necessary for the correct development and regulation of cells/organisms, aberrant splicing is now associated with an increasingly varied number of human diseases, such as neurological and developmental diseases, and cancer. Studies of splicing mechanisms and regulation are often achieved in nonhuman model organisms such as yeast. Yeasts possess homologs to many of the core spliceosome components of higher organisms, including humans, and as such yeast species are now a well-established model organism for understanding how differential splicing of transcripts can alter the phenotype of a cell or organism. Here we describe methods to investigate pre-mRNA splicing in yeast cells using modern RNA-Seq technology and bioinformatics software. Details of traditional validation methods are also described.
- Published
- 2019
31. Functional and in-silico interrogation of rare genomic variants impacting RNA splicing for the diagnosis of genomic disorders
- Author
-
Charlie Rowlands, Simon C Ramsden, Christopher O'Callaghan, Kevin Webb, Graeme C.M. Black, Robert A. Hirst, Beatriz Gomes-Silva, Andrew R. Webster, Gavin Arno, Jamie M Ellingford, Claire Hardcastle, Huw B. Thomas, William G. Newman, Jill Clayton-Smith, Raymond T. O'Keefe, Christopher J. Campbell, Glenda M. Beaman, Elizabeth A. Jones, and Nicole Gossan
- Subjects
0303 health sciences ,education.field_of_study ,business.industry ,In silico ,Alternative splicing ,Population ,Context (language use) ,Computational biology ,Biology ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,RNA splicing ,Personalized medicine ,business ,education ,Gene ,Allele frequency ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
PurposeTo develop a comprehensive analysis framework to identify pre-messenger RNA splicing mutations in the context of rare disease.MethodsWe assessed ‘variants of uncertain significance’ through six in-silico prioritization strategies. Firstly, through comparison to functional analyses, we determined the precise effect on splicing of variants identified through clinical multi-disciplinary meetings. Next, we calculated the sensitivity of in-silico prioritization strategies to distinguish known splicing mutations from common variation (>2% in allele frequency in gnomAD) within relevant disease genes. These approaches defined an accurate in-silico strategy for variant prioritization, which we retrospectively applied to a large cohort of 2783 individuals who had previously received genomic testing for rare genomic disorders. We assessed the clinical impact of such prioritization strategies alongside routine diagnostic testing strategies.ResultsWe identified 21 variants that potentially impacted splicing, and used cell based splicing assays to identify those variants which disrupted normal splicing. These findings underpinned new molecular diagnoses for 14 individuals. This process established that the use of pre-defined thresholds from a machine learning splice prediction algorithm, SpliceAI, was the most efficient method for variant prioritization, with a positive predictive value of 86%. We analysed 1,346,744 variants identified through diagnostic testing for 2783 individuals and observed that splicing variant prioritization strategies would improve clarity in clinical analysis for 15% of the individuals surveyed. Prioritized variants could provide new molecular diagnoses or provide additional support for molecular diagnosis for up to 81 individuals within our cohort.ConclusionWe present an in-silico and functional analysis framework for the assessment of variants impacting pre-messenger RNA splicing which is applicable across monogenic disorders. Incorporation of these strategies improves clarity in diagnostic reporting, increases diagnostic yield and, with the advent of targeted treatment strategies, can directly alter patient clinical management.Key HighlightsWe establish an in-silico and functional analysis framework for the incorporation of splice variant assessment into diagnostic testing that is applicable across monogenic disorders.After assessment of six distinct variant prioritization strategies, we concluded that SpliceAI was the best method to accurately identify genomic variation disrupting normal pre-mRNA splicing. We determined this through (i) functional assessment of novel ‘variants of uncertain significance’ described in this study, and (ii) calculation of sensitivity and specificity for prioritization strategies to distinguish known splicing mutations from common variants in the general population.We describe novel disease-causing variants with support from cell based functional assays which underpin autosomal recessive, autosomal dominant and X-linked Mendelian disorders. This includes variants which are deeply intronic, within the nearby splice region of canonical splice sites and variants which activate cryptic splice sites within the protein-coding regions of genes.We integrated the best performing variant prioritization strategy alongside clinical diagnostic testing for 2783 individuals referred to a well-established targeted gene panel test available through the UK National Health Service. We show that integration of such strategies will increase accuracy and clarity of diagnostic reporting, including the identification of variants which could provide new diagnoses and new carrier findings for referred individuals.Functional assessment is essential for accurate clinical assessment of variants disrupting pre-mRNA splicing. We show through cell based functional assessments that variants impacting splicing may have complex impacts on pre-mRNA splicing, which may cause multiple interpretable consequences according to ACMG guidelines.
- Published
- 2019
- Full Text
- View/download PDF
32. Author response for 'A homozygous missense variant in CHRM3 associated with familial urinary bladder disease'
- Author
-
William G. Newman, Helen M. Stuart, Adrian S. Woolf, Glenda M. Beaman, Ali Aishah, James O'Sullivan, Wee Teik Keng, Huw B. Thomas, Gabriella Galatà, Jill E. Urquhart, Katherine A. Wood, Sanjeev S. Bhaskar, and Raymond T. O'Keefe
- Subjects
medicine.medical_specialty ,business.industry ,Urology ,Medicine ,Missense mutation ,business ,Urinary bladder disease ,medicine.disease - Published
- 2019
- Full Text
- View/download PDF
33. A recurrent missense variant in HARS2 results in variable sensorineural hearing loss in three unrelated families
- Author
-
Leslie P Molina-Ramírez, Raymond T. O'Keefe, Erica H. Gerkes, Richard J.H. Smith, William G. Newman, and Leigh A M Demain
- Subjects
0301 basic medicine ,Male ,Gonadal dysgenesis ,TRANSFER-RNA SYNTHETASE ,GENE ENCODES ,Primary Ovarian Insufficiency ,CAUSE OVARIAN DYSGENESIS ,Histidine-tRNA Ligase ,0302 clinical medicine ,Missense mutation ,genetics ,Exome ,Child ,Genetics (clinical) ,Genetics ,education.field_of_study ,Homozygote ,Gonadal Dysgenesis, 46,XX ,Mitochondria ,Pedigree ,Child, Preschool ,Sensorineural hearing loss ,Female ,medicine.symptom ,Heterozygote ,Ataxia ,Hearing loss ,ovarian insufficiency ,Hearing Loss, Sensorineural ,Population ,Mutation, Missense ,rare disease ,Biology ,Article ,Amino Acyl-tRNA Synthetases ,03 medical and health sciences ,medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,education ,Alleles ,hearing loss ,Perrault syndrome ,MUTATIONS ,PERRAULT SYNDROME ,Infant ,medicine.disease ,030104 developmental biology ,Peripheral neuropathy ,030217 neurology & neurosurgery - Abstract
HARS2encodes mitochondrial histidyl-tRNA synthetase (HARS2), which links histidine to its cognate tRNA in the mitochondrial matrix. Biallelic variants inHARS2are associated with Perrault syndrome, a rare recessive condition characterized by sensorineural hearing loss in both sexes and primary ovarian insufficiency in 46,XX females. Some individuals with Perrault syndrome have a broader phenotypic spectrum with neurological features, including ataxia and peripheral neuropathy. Here, we report a recurrent variant inHARS2in association with sensorineural hearing loss. In affected individuals from three unrelated families, the variantHARS2c.1439G>A p.(Arg480His) is present as a heterozygous variant in trans to a putative pathogenic variant. The low prevalence of the alleleHARS2c.1439G>A p.(Arg480His) in the general population and its presence in three families with hearing loss, confirm the pathogenicity of this variant and illustrate the presentation of Perrault syndrome as nonsyndromic hearing loss in males and prepubertal females.
- Published
- 2019
- Full Text
- View/download PDF
34. Disease modeling of core pre-mRNA splicing factor haploinsufficiency
- Author
-
Kathryn E. Hentges, Huw B. Thomas, Weronika A. Buczek, Tracy A Briggs, Wasay Mohiuddin Shaikh Qureshi, Charlie Rowlands, Raymond T. O'Keefe, Katherine A. Wood, William G. Newman, and Simon J. Hubbard
- Subjects
introns ,ma splicing ,Gene Expression ,cell lines ,Haploinsufficiency ,exons ,Craniofacial Abnormalities ,Exon ,0302 clinical medicine ,Gene expression ,heterogeneous nuclear rna ,RNA Precursors ,ma ,rna ,genes ,Genetics (clinical) ,Regulation of gene expression ,0303 health sciences ,Gene Expression Regulation, Developmental ,General Medicine ,Exons ,Endoplasmic Reticulum Stress ,Cell biology ,Phenotype ,RNA splicing ,RNA Splicing ,Biology ,Article ,03 medical and health sciences ,Splicing factor ,Genetics ,Humans ,Craniofacial ,Molecular Biology ,Ribonucleoprotein, U5 Small Nuclear ,030304 developmental biology ,Cell Proliferation ,Sequence Analysis, RNA ,HEK 293 cells ,stress response ,Peptide Elongation Factors ,Introns ,haploinsufficiency ,HEK293 Cells ,Mutation ,gene expression ,Spliceosomes ,CRISPR-Cas Systems ,030217 neurology & neurosurgery ,Mandibulofacial Dysostosis - Abstract
The craniofacial disorder mandibulofacial dysostosis Guion-Almeida type is caused by haploinsufficiency of the U5 snRNP gene EFTUD2/SNU114. However, it is unclear how reduced expression of this core pre-mRNA splicing factor leads to craniofacial defects. Here we use a CRISPR-Cas9 nickase strategy to generate a human EFTUD2-knockdown cell line and show that reduced expression of EFTUD2 leads to diminished proliferative ability of these cells, increased sensitivity to endoplasmic reticulum (ER) stress and the mis-expression of several genes involved in the ER stress response. RNA-Seq analysis of the EFTUD2-knockdown cell line revealed transcriptome-wide changes in gene expression, with an enrichment for genes associated with processes involved in craniofacial development. Additionally, our RNA-Seq data identified widespread mis-splicing in EFTUD2-knockdown cells. Analysis of the functional and physical characteristics of mis-spliced pre-mRNAs highlighted conserved properties, including length and splice site strengths, of retained introns and skipped exons in our disease model. We also identified enriched processes associated with the affected genes, including cell death, cell and organ morphology and embryonic development. Together, these data support a model in which EFTUD2 haploinsufficiency leads to the mis-splicing of a distinct subset of pre-mRNAs with a widespread effect on gene expression, including altering the expression of ER stress response genes and genes involved in the development of the craniofacial region. The increased burden of unfolded proteins in the ER resulting from mis-splicing would exceed the capacity of the defective ER stress response, inducing apoptosis in cranial neural crest cells that would result in craniofacial abnormalities during development.
- Published
- 2019
- Full Text
- View/download PDF
35. Gene Cloning for the Analysis of Gene Expression
- Author
-
Raymond T. O'Keefe and Huw B. Thomas
- Subjects
Cloning ,Genetics ,Gibson assembly ,Chemistry ,Gene expression ,Pre-mRNA splicing ,Molecular cloning - Published
- 2019
- Full Text
- View/download PDF
36. Non-coding RNAs and disease: the classical ncRNAs make a comeback
- Author
-
Daniela Delneri, Marcin G. Fraczek, Steven Parker, Raymond T. O'Keefe, and Rogerio Alves de Almeida
- Subjects
0301 basic medicine ,RNA, Untranslated ,RNA, Mitochondrial ,Gene Expression ,Biology ,medicine.disease_cause ,Biochemistry ,Genome ,Article ,03 medical and health sciences ,medicine ,Humans ,Disease ,Genetic Predisposition to Disease ,Small nucleolar RNA ,Genetics ,Mutation ,Models, Genetic ,Genome, Human ,RNA ,Non-coding RNA ,Long non-coding RNA ,030104 developmental biology ,Transfer RNA ,Human genome - Abstract
Many human diseases have been attributed to mutation in the protein coding regions of the human genome. The protein coding portion of the human genome, however, is very small compared with the non-coding portion of the genome. As such, there are a disproportionate number of diseases attributed to the coding compared with the non-coding portion of the genome. It is now clear that the non-coding portion of the genome produces many functional non-coding RNAs and these RNAs are slowly being linked to human diseases. Here we discuss examples where mutation in classical non-coding RNAs have been attributed to human disease and identify the future potential for the non-coding portion of the genome in disease biology.
- Published
- 2016
- Full Text
- View/download PDF
37. Modelling the developmental spliceosomal craniofacial disorder Burn-McKeown syndrome using induced pluripotent stem cells
- Author
-
Huw B. Thomas, William G. Newman, Raymond T. O'Keefe, Charlie Rowlands, Julieta O’Flaherty, Katherine A. Wood, Sofia Douzgou, Susan J. Kimber, and Steven Woods
- Subjects
Gene Expression ,Apoptosis ,Deafness ,Biochemistry ,Exon ,Cell Signaling ,Neural Stem Cells ,Animal Cells ,Gene expression ,Cellular Reprogramming Techniques ,Promoter Regions, Genetic ,Induced pluripotent stem cell ,Wnt Signaling Pathway ,WNT Signaling Cascade ,Sequence Deletion ,0303 health sciences ,Multidisciplinary ,Stem Cells ,Gene Ontologies ,030305 genetics & heredity ,Wnt signaling pathway ,Gene Expression Regulation, Developmental ,Neural crest ,Cell Differentiation ,Genomics ,Exons ,Signaling Cascades ,Cell biology ,Nucleic acids ,Neural Crest ,RNA splicing ,Medicine ,Female ,Cellular Types ,Transcription Factor 7-Like 2 Protein ,Research Article ,Signal Transduction ,Heart Defects, Congenital ,Epithelial-Mesenchymal Transition ,Science ,Induced Pluripotent Stem Cells ,Biology ,Genome Complexity ,Models, Biological ,Choanal Atresia ,03 medical and health sciences ,Developmental Neuroscience ,Genetics ,Humans ,RNA, Messenger ,Craniofacial ,Ribonucleoprotein, U5 Small Nuclear ,030304 developmental biology ,Alternative splicing ,Biology and Life Sciences ,Computational Biology ,Facies ,Cell Biology ,Genome Analysis ,Introns ,Clone Cells ,Alternative Splicing ,RNA processing ,Cellular Neuroscience ,Face ,Spliceosomes ,RNA ,RNA Splice Sites ,Head ,Developmental Biology ,Neuroscience - Abstract
The craniofacial developmental disorder Burn-McKeown Syndrome (BMKS) is caused by biallelic variants in the pre-messenger RNA splicing factor gene TXNL4A/DIB1. The majority of affected individuals with BMKS have a 34 base pair deletion in the promoter region of one allele of TXNL4A combined with a loss-of-function variant on the other allele, resulting in reduced TXNL4A expression. However, it is unclear how reduced expression of this ubiquitously expressed spliceosome protein results in craniofacial defects during development. Here we reprogrammed peripheral mononuclear blood cells from a BMKS patient and her unaffected mother into induced pluripotent stem cells (iPSCs) and differentiated the iPSCs into induced neural crest cells (iNCCs), the key cell type required for correct craniofacial development. BMKS patient-derived iPSCs proliferated more slowly than both mother- and unrelated control-derived iPSCs, and RNA-Seq analysis revealed significant differences in gene expression and alternative splicing. Patient iPSCs displayed defective differentiation into iNCCs compared to maternal and unrelated control iPSCs, in particular a delay in undergoing an epithelial-to-mesenchymal transition (EMT). RNA-Seq analysis of differentiated iNCCs revealed widespread gene expression changes and mis-splicing in genes relevant to craniofacial and embryonic development that highlight a dampened response to WNT signalling, the key pathway activated during iNCC differentiation. Furthermore, we identified the mis-splicing of TCF7L2 exon 4, a key gene in the WNT pathway, as a potential cause of the downregulated WNT response in patient cells. Additionally, mis-spliced genes shared common sequence properties such as length, splice site strengths and sequence motifs, suggesting that splicing of particular subsets of genes is particularly sensitive to changes in TXNL4A expression. Together, these data provide the first insight into how reduced TXNL4A expression in BMKS patients might compromise splicing and NCC function, resulting in defective craniofacial development in the embryo.
- Published
- 2020
- Full Text
- View/download PDF
38. Advanced Methods for the Analysis of Altered Pre-mRNA Splicing in Yeast and Disease
- Author
-
Huw B. Thomas and Raymond T. O'Keefe
- Subjects
0303 health sciences ,Spliceosome ,ved/biology ,030305 genetics & heredity ,Alternative splicing ,ved/biology.organism_classification_rank.species ,RNA ,Computational biology ,Biology ,Phenotype ,Yeast ,03 medical and health sciences ,RNA splicing ,Model organism ,Organism ,030304 developmental biology - Abstract
Splicing of pre-messenger RNA (pre-mRNA) transcripts is a fundamental process in all eukaryotes that provides a mechanism of increasing the proteomic diversity within a cell that can be tightly regulated in a dynamic manner. While constitutive and alternative splicing are necessary for the correct development and regulation of cells/organisms, aberrant splicing is now associated with an increasingly varied number of human diseases, such as neurological and developmental diseases, and cancer. Studies of splicing mechanisms and regulation are often achieved in nonhuman model organisms such as yeast. Yeasts possess homologs to many of the core spliceosome components of higher organisms, including humans, and as such yeast species are now a well-established model organism for understanding how differential splicing of transcripts can alter the phenotype of a cell or organism. Here we describe methods to investigate pre-mRNA splicing in yeast cells using modern RNA-Seq technology and bioinformatics software. Details of traditional validation methods are also described.
- Published
- 2019
- Full Text
- View/download PDF
39. A Known Pathogenic Variant in the Essential Mitochondrial Translation Gene RMND1 Causes a Perrault-Like Syndrome with Renal Defects
- Author
-
S.S. Bhaskhar, Raymond T. O'Keefe, James O'Sullivan, Leigh A M Demain, Diana Antunes, and William G. Newman
- Subjects
0301 basic medicine ,Letter ,Mitochondrial translation ,Hearing Loss, Sensorineural ,HDE GEN ,Cell Cycle Proteins ,030105 genetics & heredity ,Mitochondrion ,Whole Exome Sequencing ,03 medical and health sciences ,Exome Sequencing ,Genetics ,Medicine ,Humans ,Genetic Predisposition to Disease ,Cell Cycle Protein ,Gene ,Genetics (clinical) ,Exome sequencing ,business.industry ,Gonadal Dysgenesis, 46,XX ,Mitochondria ,030104 developmental biology ,Female ,Kidney Diseases ,business - Abstract
Submitted by Dulce Barreto (mdulce.barreto@chlc.min-saude.pt) on 2020-05-22T13:46:36Z No. of bitstreams: 1 Clin Genet 2018_1.pdf: 281862 bytes, checksum: 5c561ef864326f17ad872bf33ed8e872 (MD5) Made available in DSpace on 2020-05-22T13:46:36Z (GMT). No. of bitstreams: 1 Clin Genet 2018_1.pdf: 281862 bytes, checksum: 5c561ef864326f17ad872bf33ed8e872 (MD5) Previous issue date: 2018 info:eu-repo/semantics/publishedVersion
- Published
- 2018
- Full Text
- View/download PDF
40. A homozygous variant in mitochondrial RNase P subunit PRORP is associated with Perrault syndrome characterized by hearing loss and primary ovarian insufficiency
- Author
-
Jill E. Urquhart, Zeev Blumenfeld, Emma M. Jenkinson, Johannes Zschocke, Albert Amberger, Walter Rossmanith, James O'Sullivan, William G. Newman, Thomas B. Friedman, Kyle Thompson, Irit Hochberg, Wyatt W. Yue, Leigh Ann Mary Demain, Andrea J. Deutschmann, Kevin J. Munro, Stephanie Oerum, Sergey Yalonetsky, Nada Al-Sheqaih, Sandra Demetz, Raymond T. O'Keefe, Robert W. Taylor, Sanjeev S. Bhaskar, Inna A. Belyantseva, Melanie Barzik, and Simon G. Williams
- Subjects
Genetics ,0303 health sciences ,Messenger RNA ,Mitochondrial translation ,RNase P ,Wild type ,TRNA processing ,Biology ,medicine.disease ,Molecular biology ,03 medical and health sciences ,0302 clinical medicine ,Transfer RNA ,medicine ,Missense mutation ,Sensorineural hearing loss ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Perrault syndrome is a rare autosomal recessive condition characterised by sensorineural hearing loss in both sexes and primary ovarian insufficiency in 46 XX, females. It is genetically heterogeneous with biallelic variants in six genes identified to date (HSD17B4, HARS2, LARS2, CLPP, C10orf2 and ERAL1). Most genes possessing variants associated with Perrault syndrome are involved in mitochondrial translation. We describe a consanguineous family with three affected individuals homozygous for a novel missense variant c.1454C>T; p.(Ala485Val) in KIAA0391, encoding proteinaceous RNase P (PRORP), the metallonuclease subunit of the mitochondrial RNase P complex, responsible for the 5’-end processing of mitochondrial precursor tRNAs. In RNase P activity assays, RNase P complexes containing the PRORP disease variant produced ~35-45% less 5’-processed tRNA than wild type PRORP. Consistently, the accumulation of unprocessed polycistronic mitochondrial transcripts was observed in patient dermal fibroblasts, leading to an observable loss of steady-state levels of mitochondrial oxidative phosphorylation components. Expression of wild type KIAA0391 in patient fibroblasts rescued tRNA processing. Immunohistochemistry analyses of the auditory sensory epithelium from postnatal and adult mouse inner ear showed a high level of PRORP in the efferent synapses and nerve fibres of hair cells, indicating a possible mechanism for the sensorineural hearing loss observed in affected individuals. We have identified a variant in an additional gene associated with Perrault syndrome. With the identification of this disease-causing variant in KIAA0391, reduced function of each of the three subunits of mitochondrial RNase P have now been associated with distinct clinical presentations.Author SummaryPerrault syndrome is a rare genetic condition which results in hearing loss in both sexes and ovarian dysfunction in females. Perrault syndrome may also cause neurological symptoms in some patients. Here, we present the features and genetic basis of the condition in three sisters affected by Perrault syndrome. The sisters did not have pathogenic variants in any of the genes previously associated with Perrault syndrome. We identified a change in the gene KIAA0391, encoding PRORP, a subunit of the mitochondrial RNase P complex. Mitochondrial RNase P is a key enzyme in RNA processing in mitochondria. Impaired RNA processing reduces protein production in mitochondria, which we observed in patient cells along with high levels of unprocessed RNA. When we expressed wild type PRORP in patient cells, the RNA processing improved. We also investigated PRORP localisation in the mouse inner ear and found high levels in the synapses and nerve fibers that transmit sound. It may be that disruption of RNA processing in the mitochondria of these cells causes hearing loss in this family.
- Published
- 2017
- Full Text
- View/download PDF
41. Corrigendum: Mutations in SNORD118 cause the cerebral microangiopathy leukoencephalopathy with calcifications and cysts
- Author
-
Rosaline Caumes, Andrea Berger, John Stone, Roberta Battini, Charles Marques Lourenço, Gerardine Quaghebeur, Nicholas A. Fletcher, Elliott H. Sherr, Marjo S. van der Knaap, Sanjeev S. Bhaskar, Marco Henneke, Kanaga R. Sinnathuray, Pierre Landrieu, James O'Sullivan, Calvin Soh, Himanshu Goel, Patrick Ferreira, Katrin Õunap, Cheryl Hemingway, Laurence C. Goosey, Graham D. Pavitt, Hilde Van Esch, Prab Prabhakar, Yoann Rose, Jill E. Urquhart, Yanick J. Crow, Gabriela M. Baerlocher, Simon G. Williams, Mathieu P Rodero, Ram L. Kumar, Emma M. Jenkinson, Gillian I. Rice, Sarju G. Mehta, Adeline Vanderver, Timothy J. Malpas, Christopher J. Kershaw, Imke Metz, Edward Blair, Paul R. Kasher, Imelda Hughes, Patrick Revy, David Cassiman, Duccio Maria Cordelli, Angela Barnicoat, Andrea Whitney, Janice E. Brunstrom-Hernandez, Axel Panzer, Kristin W. Barañano, Liesbeth De Waele, Geraldine Aubert, Raphael Schiffmann, H. Stewart, Monika Haubitz, Rosalind J. Jefferson, Alan Fryer, Anthony Oojageer, Carolina Uggenti, John H. Livingston, Sam Griffiths-Jones, Christine E. G. van Mol, Johannes A. Buckard, Raymond T. O'Keefe, Lieven Lagae, Emma Wakeling, Sakkubai Naidu, Alex J. Fay, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques (LCBPT - UMR 8601), Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Manchester Centre for Genomic Medicine (MCGM), Manchester Academic Health Science Centre (MAHSC), University of Manchester [Manchester]-University of Manchester [Manchester]-Faculty of Biology, Medicine and Health [Manchester, UK], University of Manchester [Manchester]-Manchester University NHS Foundation Trust (MFT)-St Mary's Hospital Manchester, Laboratory of neurogenetics and neuroinflammation (Equipe Inserm U1163), Imagine - Institut des maladies génétiques (IMAGINE - U1163), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Faculty of Biology, Medicine and Health [Manchester, UK], University of Manchester [Manchester], Department of Medical Genetics, and HMNC Brain Health
- Subjects
0303 health sciences ,Pathology ,medicine.medical_specialty ,[SDV]Life Sciences [q-bio] ,Biology ,medicine.disease ,Article ,3. Good health ,Leukoencephalopathy ,03 medical and health sciences ,0302 clinical medicine ,Cerebral microangiopathy ,Genetics ,medicine ,610 Medicine & health ,030217 neurology & neurosurgery ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology - Abstract
Although ribosomes are ubiquitously expressed and essential for life, recent data indicate that monogenic causes of ribosomal dysfunction can confer a remarkable degree of specificity in terms of human disease phenotype. Box C/D small nucleolar RNAs (snoRNAs) are evolutionarily conserved non-protein encoding RNAs involved in ribosome biogenesis. Here we show that biallelic mutations in the gene SNORD118, encoding the box C/D snoRNA U8, cause the cerebral microangiopathy leukoencephalopathy with calcifications and cysts (LCC), presenting at any age from early childhood to late adulthood. These mutations affect U8 expression, processing and protein binding and thus implicate U8 as essential in cerebral vascular homeostasis.
- Published
- 2017
- Full Text
- View/download PDF
42. Remodeling of U2-U6 snRNA helix I during pre-mRNA splicing by Prp16 and the NineTeen Complex protein Cwc2
- Author
-
Jayalath P.D. Ruckshanthi, Rogerio Alves de Almeida, Rebecca Hogg, and Raymond T. O'Keefe
- Subjects
Adenosine Triphosphatases ,Genetics ,RNA Splicing Factors ,Spliceosome ,Saccharomyces cerevisiae Proteins ,RNA Splicing ,Intron ,RNA-Binding Proteins ,Prp24 ,Biology ,Cell biology ,SR protein ,Polypyrimidine tract ,RNA, Small Nuclear ,Mutation ,RNA splicing ,RNA Precursors ,RNA ,Nucleic Acid Conformation ,snRNP ,RNA, Messenger ,RNA Helicases - Abstract
Removal of intron regions from pre-messenger RNA (pre-mRNA) requires spliceosome assembly with pre-mRNA, then subsequent spliceosome remodeling to allow activation for the two steps of intron removal. Spliceosome remodeling is carried out through the action of DExD/H-box ATPases that modulate RNA-RNA and protein-RNA interactions. The ATPase Prp16 remodels the spliceosome between the first and second steps of splicing by catalyzing release of first step factors Yju2 and Cwc25 as well as destabilizing U2-U6 snRNA helix I. How Prp16 destabilizes U2-U6 helix I is not clear. We show that the NineTeen Complex (NTC) protein Cwc2 displays genetic interactions with the U6 ACAGAGA, the U6 internal stem loop (ISL) and the U2-U6 helix I, all RNA elements that form the spliceosome active site. We find that one function of Cwc2 is to stabilize U2-U6 snRNA helix I during splicing. Cwc2 also functionally cooperates with the NTC protein Isy1/NTC30. Mutation in Cwc2 can suppress the cold sensitive phenotype of the prp16-302 mutation indicating a functional link between Cwc2 and Prp16. Specifically the prp16-302 mutation in Prp16 stabilizes Cwc2 interactions with U6 snRNA and destabilizes Cwc2 interactions with pre-mRNA, indicating antagonistic functions of Cwc2 and Prp16. We propose that Cwc2 is a target for Prp16-mediated spliceosome remodeling during pre-mRNA splicing.
- Published
- 2014
- Full Text
- View/download PDF
43. <scp>T</scp>he U5 sn<scp>RNA</scp>internal loop 1 is a platform for Brr2, Snu114 and Prp8 protein binding during U5 sn<scp>RNP</scp>assembly
- Author
-
Raymond T. O'Keefe, Jayalath P.D. Ruckshanthi, Verity Nancollis, and Lily Novak Frazer
- Subjects
U5 snRNP ,Spliceosome ,Saccharomyces cerevisiae Proteins ,Ribonucleoprotein, U4-U6 Small Nuclear ,Saccharomyces cerevisiae ,SPLICEOSOME ,Brr2 ,Prp24 ,Plasma protein binding ,Prp8 ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,RNA, Small Nuclear ,PRE-mRNA SPLICING ,snRNP ,Molecular Biology ,Ribonucleoprotein, U5 Small Nuclear ,030304 developmental biology ,SACCHAROMYCES CEREVISIAE ,Genetics ,0303 health sciences ,biology ,Small Nuclear Ribonucleoprotein Particle ,Intron ,Articles ,Cell Biology ,Ribonucleoproteins, Small Nuclear ,biology.organism_classification ,Cell biology ,Mutation ,Spliceosomes ,Nucleic Acid Conformation ,Snu114 ,RNA Helicases ,030217 neurology & neurosurgery ,Small nuclear RNA ,Protein Binding - Abstract
The U5 small nuclear ribonucleoprotein particle (snRNP) forms the heart of the spliceosome which is required for intron removal from pre-mRNA. The proteins Prp8, Snu114 and Brr2 all assemble with the U5 small nuclear RNA (snRNA) to produce the U5 snRNP. Successful assembly of the U5 snRNP, then incorporation of this snRNP into the U4/U6.U5 tri-snRNP and the spliceosome, is essential for producing an active spliceosome. We have investigated the requirements for Prp8, Snu114 and Brr2 association with the U5 snRNA to form the U5 snRNP in yeast. Mutations were constructed in the highly conserved loop 1 and internal loop 1 (IL1) of the U5 snRNA and their function assessed in vivo. The influence of these U5 mutations on association of Prp8, Snu114 and Brr2 with the U5 snRNA were then determined. U5 snRNA loop 1 and both sides of IL1 in U5 were important for association of Prp8, Snu114 and Brr2 with the U5 snRNA. Mutations in the 3′ side of U5 IL1 resulted in the greatest reduction of Prp8, Snu114 and Brr2 association with the U5 snRNA. Genetic screening of brr2 and U5 snRNA mutants revealed synthetic lethal interactions between alleles in Brr2 and the 3′ side of U5 snRNA IL1 which reflects reduced association between Brr2 and U5 IL1. We propose that the U5 snRNA IL1 is a platform for protein binding and is required for Prp8, Brr2 and Snu114 association with the U5 snRNA to form the U5 snRNP. J. Cell. Biochem. 114: 2770–2784, 2013. © 2013 The Authors. Journal of Cellular Biochemistry Published by Wiley Periodicals Inc.
- Published
- 2013
- Full Text
- View/download PDF
44. Mutations in SNORD118 cause the cerebral microangiopathy leukoencephalopathy with calcifications and cysts
- Author
-
Elliott H. Sherr, Edward Blair, Charles Marques Lourenço, James O'Sullivan, Imke Metz, Paul R. Kasher, Gabriela M. Baerlocher, Adeline Vanderver, David Cassiman, Himanshu Goel, Nicholas A. Fletcher, Patrick Ferreira, Patrick Revy, Emma Wakeling, Ram L. Kumar, Lieven Lagae, Christopher J. Kershaw, Pierre Landrieu, Andrea Whitney, Calvin Soh, Christine E. G. van Mol, Sakkubai Naidu, John H. Livingston, Geraldine Aubert, H. Stewart, Laurence C. Goosey, Liesbeth De Waele, Kristin W. Barañano, Rosalind J. Jefferson, Axel Panzer, Gerardine Quaghebeur, Raphael Schiffmann, Yanick J. Crow, Hilde Van Esch, Raymond T. O'Keefe, Jill E. Urquhart, Alan Fryer, Mathieu P Rodero, Alex J. Fay, Monika Haubitz, Andrea Berger, Johannes A. Buckard, Cheryl Hemingway, Angela Barnicoat, Sam Griffiths-Jones, Duccio Maria Cordelli, Imelda Hughes, Katrin Õunap, Graham D. Pavitt, Roberta Battini, Yoann Rose, Marjo S. van der Knaap, Sanjeev S. Bhaskar, John Stone, Gillian I. Rice, Marco Henneke, Kanaga R. Sinnathuray, Emma M. Jenkinson, Timothy J. Malpas, Simon G. Williams, Anthony Oojageer, Carolina Uggenti, Rosaline Caumes, Prab Prabhakar, Sarju G. Mehta, Janice E. Brunstrom-Hernandez, Manchester Centre for Genomic Medicine (MCGM), Manchester Academic Health Science Centre (MAHSC), University of Manchester [Manchester]-University of Manchester [Manchester]-Faculty of Biology, Medicine and Health [Manchester, UK], University of Manchester [Manchester]-Manchester University NHS Foundation Trust (MFT)-St Mary's Hospital Manchester, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques (LCBPT - UMR 8601), Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Imagine - Institut des maladies génétiques (IMAGINE - U1163), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), University of Manchester [Manchester], Department of Medical Genetics, HMNC Brain Health, Other departments, Jenkinson, Emma M., Rodero, Mathieu P., Kasher, Paul R., Uggenti, Carolina, Oojageer, Anthony, Goosey, Laurence C., Rose, Yoann, Kershaw, Christopher J., Urquhart, Jill E., Williams, Simon G., Bhaskar, Sanjeev S., O'Sullivan, Jame, Baerlocher, Gabriela M., Haubitz, Monika, Aubert, Geraldine, Barañano, Kristin W., Barnicoat, Angela J., Battini, Roberta, Berger, Andrea, Blair, Edward M., Brunstrom-Hernandez, Janice E., Buckard, Johannes A., Cassiman, David M., Caumes, Rosaline, Cordelli, Duccio M., De Waele, Liesbeth M., Fay, Alexander J., Ferreira, Patrick, Fletcher, Nicholas A., Fryer, Alan E., Goel, Himanshu, Hemingway, Cheryl A., Henneke, Marco, Hughes, Imelda, Jefferson, Rosalind J., Kumar, Ram, Lagae, Lieven, Landrieu, Pierre G., Lourenço, Charles M., Malpas, Timothy J., Mehta, Sarju G., Metz, Imke, Naidu, Sakkubai, Õunap, Katrin, Panzer, Axel, Prabhakar, Prab, Quaghebeur, Gerardine, Schiffmann, Raphael, Sherr, Elliott H., Sinnathuray, Kanaga R., Soh, Calvin, Stewart, Helen S., Stone, John, Van Esch, Hilde, Van Mol, Christine E. G., Vanderver, Adeline, Wakeling, Emma L., Whitney, Andrea, Pavitt, Graham D., Griffiths-Jones, Sam, Rice, Gillian I., Revy, Patrick, Van Der Knaap, Marjo S., Livingston, John H., O'Keefe, Raymond T., Crow, Yanick J., Pediatric surgery, and Amsterdam Neuroscience - Cellular & Molecular Mechanisms
- Subjects
0301 basic medicine ,Male ,Genetic Linkage ,[SDV]Life Sciences [q-bio] ,Ribosome biogenesis ,medicine.disease_cause ,Leukoencephalopathy ,Leukoencephalopathie ,Cohort Studies ,0302 clinical medicine ,Leukoencephalopathies ,Exome ,Small nucleolar RNA ,610 Medicine & health ,Child ,ComputingMilieux_MISCELLANEOUS ,Genetics ,Mutation ,Genome ,Cysts ,Calcinosis ,Middle Aged ,Phenotype ,Child, Preschool ,Calcinosi ,Female ,Sequence Analysis ,Human ,Adult ,Adolescent ,Biology ,Chromosomes ,Cell Line ,03 medical and health sciences ,Young Adult ,medicine ,RNA, Small Nucleolar ,Humans ,Preschool ,Gene ,Small Nucleolar ,Genome, Human ,Pair 17 ,RNA ,Infant ,Sequence Analysis, DNA ,DNA ,Cerebral Small Vessel Diseases ,Chromosomes, Human, Pair 17 ,medicine.disease ,Cerebral Small Vessel Disease ,030104 developmental biology ,Cyst ,Cohort Studie ,030217 neurology & neurosurgery - Abstract
Although ribosomes are ubiquitous and essential for life, recent data indicate that monogenic causes of ribosomal dysfunction can confer a remarkable degree of specificity in terms of human disease phenotype. Box C/D small nucleolar RNAs (snoRNAs) are evolutionarily conserved non-protein-coding RNAs involved in ribosome biogenesis. Here we show that biallelic mutations in the gene SNORD118, encoding the box C/D snoRNA U8, cause the cerebral microangiopathy leukoencephalopathy with calcifications and cysts (LCC), presenting at any age from early childhood to late adulthood. These mutations affect U8 expression, processing and protein binding and thus implicate U8 as essential in cerebral vascular homeostasis.
- Published
- 2016
- Full Text
- View/download PDF
45. The function of the NineTeen Complex (NTC) in regulating spliceosome conformations and fidelity during pre-mRNA splicing
- Author
-
Joanne C. McGrail, Raymond T. O'Keefe, and Rebecca Hogg
- Subjects
Genetics ,RNA Splicing Factors ,Spliceosome ,Saccharomyces cerevisiae Proteins ,RNA Splicing ,Molecular Conformation ,Intron ,Saccharomyces cerevisiae ,Biology ,Models, Biological ,Biochemistry ,Article ,Yeast ,Cell biology ,Minor spliceosome ,RNA splicing ,RNA Precursors ,Spliceosomes ,Humans ,snRNP ,Function (biology) - Abstract
The NineTeen Complex (NTC) of proteins associates with the spliceosome during pre-mRNA splicing and is essential for both steps of intron removal. The NTC and other NTC-associated proteins are recruited to the spliceosome where they participate in regulating the formation and progression of essential spliceosome conformations required for the two steps of splicing. It is now clear that the NTC is an integral component of active spliceosomes from yeast to humans and provides essential support for the spliceosomal snRNPs (small nuclear ribonucleoproteins). In the present article, we discuss the identification and characterization of the yeast NTC and review recent work in yeast that supports the essential role for this complex in the regulation and fidelity of splicing.
- Published
- 2010
- Full Text
- View/download PDF
46. The RNA binding protein Cwc2 interacts directly with the U6 snRNA to link the nineteen complex to the spliceosome during pre-mRNA splicing
- Author
-
Joanne C. McGrail, Raymond T. O'Keefe, and Andre Krause
- Subjects
Spliceosome ,Saccharomyces cerevisiae Proteins ,RNA Splicing ,Amino Acid Motifs ,Molecular Sequence Data ,Prp24 ,RNA-binding protein ,Saccharomyces cerevisiae ,Biology ,03 medical and health sciences ,Gene Expression Regulation, Fungal ,RNA, Small Nuclear ,Genetics ,RNA Precursors ,snRNP ,Amino Acid Sequence ,RNA, Messenger ,030304 developmental biology ,0303 health sciences ,Binding Sites ,urogenital system ,030302 biochemistry & molecular biology ,fungi ,Intron ,RNA ,RNA-Binding Proteins ,Zinc Fingers ,Molecular biology ,Cell biology ,RNA splicing ,Mutation ,Spliceosomes ,Small nuclear RNA - Abstract
Intron removal during pre-messenger RNA (pre-mRNA) splicing involves arrangement of snRNAs into conformations that promote the two catalytic steps. The Prp19 complex [nineteen complex (NTC)] can specify U5 and U6 snRNA interactions with pre-mRNA during spliceosome activation. A candidate for linking the NTC to the snRNAs is the NTC protein Cwc2, which contains motifs known to bind RNA, a zinc finger and RNA recognition motif (RRM). In yeast cells mutation of either the zinc finger or RRM destabilize Cwc2 and are lethal. Yeast cells depleted of Cwc2 accumulate pre-mRNA and display reduced levels of U1, U4, U5 and U6 snRNAs. Cwc2 depletion also reduces U4/U6 snRNA complex levels, as found with depletion of other NTC proteins, but without increase in free U4. Purified Cwc2 displays general RNA binding properties and can bind both snRNAs and pre-mRNA in vitro. A Cwc2 RRM fragment alone can bind RNA but with reduced efficiency. Under splicing conditions Cwc2 can associate with U2, U5 and U6 snRNAs, but can only be crosslinked directly to the U6 snRNA. Cwc2 associates with U6 both before and after the first step of splicing. We propose that Cwc2 links the NTC to the spliceosome during pre-mRNA splicing through the U6 snRNA.
- Published
- 2009
47. Analysis of pre-mRNA and pre-rRNA processing factor Snu13p structure and mutants
- Author
-
Jordi Bella, Raymond T. O'Keefe, Helen C. Dobbyn, Paul A. McEwan, Lily Novak-Frazer, and Andre Krause
- Subjects
Models, Molecular ,Saccharomyces cerevisiae Proteins ,Biophysics ,RNA ,Prp24 ,RNA-binding protein ,Saccharomyces cerevisiae ,Cell Biology ,Biology ,Ribonucleoproteins, Small Nuclear ,Heterogeneous ribonucleoprotein particle ,Non-coding RNA ,Biochemistry ,Molecular biology ,Cell biology ,RNA, Ribosomal ,Mutation ,RNA Precursors ,snRNP ,Signal recognition particle RNA ,RNA, Messenger ,Cloning, Molecular ,Molecular Biology ,Small nuclear RNA - Abstract
Snu13p is a Saccharomyces cerevisiae protein essential for pre-messenger RNA splicing and pre-ribosomal RNA processing. Snu13p binds U4 snRNA of the spliceosome and box C/D snoRNAs of the pre-ribosomal RNA processing machinery to induce assembly of each ribonucleoprotein complex. Here, we present structural and biochemical analysis of Snu13p. The crystal structure of Snu13p reveals a region of the protein which could be important for protein interaction during ribonucleoprotein assembly. Using the structure of Snu13p we have designed the first temperature-sensitive mutants in Snu13p, L67W and I102A. Wild-type and mutant Snu13p proteins were assayed for binding to U4 snRNA and U3 snoRNA. Both temperature-sensitive mutants displayed significantly reduced RNA binding compared to wild-type protein. As the temperature-sensitive mutations are not in the known RNA binding region of Snu13p this indicates that these mutants indirectly influence the RNA binding properties of Snu13p. This work provides insight into Snu13p function during ribonucleoprotein assembly.
- Published
- 2007
- Full Text
- View/download PDF
48. The NineTeen Complex (NTC) and NTC-associated proteins as targets for spliceosomal ATPase action during pre-mRNA splicing
- Author
-
Raymond T. O'Keefe and Rogerio Alves de Almeida
- Subjects
Spliceosome ,Prp16 ,Saccharomyces cerevisiae Proteins ,RNA helicase ,Mature messenger RNA ,RNA Splicing ,Prp19 ,Brr2 ,RNA-binding protein ,Saccharomyces cerevisiae ,Biology ,DEAD-box RNA Helicases ,PremRNA splicing ,Minor spliceosome ,Prp2 ,RNA Precursors ,Humans ,ATPase ,Molecular Biology ,Genetics ,Adenosine Triphosphatases ,Intron ,RNA ,RNA-Binding Proteins ,Point-of-Views ,Cell Biology ,Exons ,Introns ,Cell biology ,Prp43 ,Polypyrimidine tract ,NineTeen Complex ,RNA splicing ,Spliceosomes ,Cwc2 - Abstract
Pre-mRNA splicing is an essential step in gene expression that removes intron sequences efficiently and accurately to produce a mature mRNA for translation. It is the large and dynamic RNA-protein complex called the spliceosome that catalyzes intron removal. To carry out splicing the spliceosome not only needs to assemble correctly with the pre-mRNA but the spliceosome requires extensive remodelling of its RNA and protein components to execute the 2 steps of intron removal. Spliceosome remodelling is achieved through the action of ATPases that target both RNA and proteins to produce spliceosome conformations competent for each step of spliceosome activation, catalysis and disassembly. An increasing amount of research has pointed to the spliceosome associated NineTeen Complex (NTC) of proteins as targets for the action of a number of the spliceosomal ATPases during spliceosome remodelling. In this point-of-view article we present the latest findings on the changes in the NTC that occur following ATPase action that are required for spliceosome activation, catalysis and disassembly. We proposed that the NTC is one of the main targets of ATPase action during spliceosome remodelling required for pre-mRNA splicing.
- Published
- 2015
49. Mutation in the U2 snRNA influences exon interactions of U5 snRNA loop 1 during pre-mRNA splicing
- Author
-
Elaine M Tatum, Raymond T. O'Keefe, and Joanne C. McGrail
- Subjects
Genetics ,Base Sequence ,General Immunology and Microbiology ,RNA Splicing ,General Neuroscience ,Molecular Sequence Data ,Alternative splicing ,Exonic splicing enhancer ,Intron ,Prp24 ,Exons ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Exon ,Splicing factor ,RNA, Small Nuclear ,Yeasts ,Mutation ,RNA splicing ,RNA Precursors ,Nucleic Acid Conformation ,snRNP ,Molecular Biology - Abstract
The U2 and U6 snRNAs contribute to the catalysis of intron removal while U5 snRNA loop 1 holds the exons for ligation during pre-mRNA splicing. It is unclear how different exons are positioned precisely with U5 loop 1. Here, we investigate the role of U2 and U6 in positioning the exons with U5 loop 1. Reconstitution in vitro of spliceosomes with mutations in U2 allows U5-pre-mRNA interactions before the first step of splicing. However, insertion in U2 helix Ia disrupts U5-exon interactions with the intron lariat-3' exon splicing intermediate. Conversely, U6 helix Ia insertions prevent U5-pre-mRNA interactions before the first step of splicing. In vivo, synthetic lethal interactions have been identified between U2 insertion and U5 loop 1 insertion mutants. Additionally, analysis of U2 insertion mutants in vivo reveals that they influence the efficiency, but not the accuracy of splicing. Our data suggest that U2 aligns the exons with U5 loop 1 for ligation during the second step of pre-mRNA splicing.
- Published
- 2006
- Full Text
- View/download PDF
50. The apolipoprotein B mRNA editing complex performs a multifunctional cycle and suppresses nonsense-mediated decay
- Author
-
Raymond T. O'Keefe, James Scott, Naveenan Navaratnam, Maria Tzimina, Angelika Somasekaram, Adam Jarmuz, Ann Chester, and Jane Gisbourne
- Subjects
Cytoplasm ,Apolipoprotein B ,APOBEC-1 Deaminase ,Nuclear Localization Signals ,Nonsense-mediated decay ,Biology ,digestive system ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Cytidine Deaminase ,Two-Hybrid System Techniques ,Animals ,Humans ,RNA, Messenger ,Nuclear export signal ,Molecular Biology ,Cell Nucleus ,Messenger RNA ,General Immunology and Microbiology ,General Neuroscience ,APOBEC1 ,Biological Transport ,Articles ,Cytidine deaminase ,Molecular biology ,MRNA editing complex ,digestive system diseases ,RNA editing ,biology.protein ,RNA Editing - Abstract
The C to U editing of apolipoprotein B (apoB) mRNA is mediated by a minimal complex composed of an RNA-binding cytidine deaminase (APOBEC1) and a complementing specificity factor (ACF). This editing generates a premature termination codon and a truncated open reading frame. We demonstrate that the APOBEC1–ACF holoenzyme mediates a multifunctional cycle. The atypical APOBEC1 nuclear localization signal is involved in RNA binding and is used to import ACF into the nucleus as cargo. APOBEC1 alone induces nonsense-mediated decay (NMD). The APOBEC1–ACF complex edits and remains associated with the edited RNA to protect it from NMD. The APOBEC1 nuclear export signal is involved in the export of ACF and the edited apoB mRNA together, to the site of translation.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.