Functional Consequences of Setd1a Haploinsufficiency: from Gestation to Behaviour

Total Page:16

File Type:pdf, Size:1020Kb

Functional Consequences of Setd1a Haploinsufficiency: from Gestation to Behaviour Functional consequences of Setd1a haploinsufficiency: from gestation to behaviour by Matthew Lee Bosworth A thesis submitted for the degree of Doctor of Philosophy September 2019 Declarations Statement 1 This thesis is being submitted in partial fulfilment of the requirements for the degree of Doctor of Philosophy. ____________________________ (Matthew Bosworth) Date 24/09/2019 Statement 2 This work has not been submitted in substance for any other degree or award at this or any other university or place of learning, nor is it being submitted concurrently for any other degree or award (outside of any formal collaboration agreement between the University and a partner organisation) ____________________________ (Matthew Bosworth) Date 24/09/2019 Statement 3 I hereby give consent for my thesis, if accepted, to be available in the University’s Open Access repository (or, where approved, to be available in the University's library and for inter-library loan), and for the title and summary to be made available to outside organisations, subject to the expiry of a University-approved bar on access if applicable. ____________________________ (Matthew Bosworth) Date 24/09/2019 Declaration This thesis is the result of my own independent work, except where otherwise stated, and the views expressed are my own. Other sources are acknowledged by explicit references. The thesis has not been edited by a third party beyond what is permitted by Cardiff University's Use of Third Party Editors by Research Degree Students Procedure. ____________________________ (Matthew Bosworth) Date 24/09/2019 i ii Summary Advances in psychiatric genetics are providing opportunities to investigate underlying pathogenic mechanisms. Rare loss of function (LoF) variants that have large effects on risk are of particular interest because they unequivocally implicate LoF of a single gene and are expected to have prominent phenotypic effects. The first LoF variants to be identified for schizophrenia were in the SETD1A gene. SETD1A catalyses methylation of lysine residue 4 on histone 3 (H3K4) and its pathogenic role is consistent with convergent evidence implicating disrupted H3K4 methylation in schizophrenia and other neurodevelopmental disorders. However, understanding of the biological mechanisms underlying the association between SETD1A LoF and psychopathology is lacking. This thesis investigated the functional consequences of Setd1a haploinsufficiency using a mouse model. Setd1a haploinsufficiency resulted in modest transcriptomic changes in the developing mouse brain that were enriched for mitochondrial annotations (Chapter 3). However, there was no enrichment for schizophrenia common variant association. Additionally, placental weight was reduced in Setd1a+/- mice and sexually dimorphic changes in placental gene expression and postnatal growth were observed in Setd1a+/- males but not females (Chapter 4). Behavioural phenotyping revealed that constitutive Setd1a haploinsufficiency caused heightened emotional reactivity and aberrant sensorimotor gating (Chapter 5). The effects on sensorimotor gating were robust and could not be rescued by established antipsychotics (haloperidol and risperidone) (Chapter 6). However, male (but not female) Setd1a+/- mice showed an insensitivity to the startle-inhibiting effects of risperidone, potentially indicating 5-HT2A receptor dysfunction. Finally, behavioural phenotyping of a conditional knockout with Setd1a haploinsufficiency constrained to the nervous system revealed evidence for a degree of convergence with the constitutive model (Chapter 7). In conclusion, this thesis provides novel insights into the diverse effects of Setd1a haploinsufficiency. These findings warrant further investigation to establish the pathophysiological relevance of effects on the developing brain, placenta, and adult brain function. iii iv List of Contents Section Page Chapter 1: General Introduction 1 1.1. Unmet clinical need of schizophrenia 1 1.2. Neurobiology of schizophrenia 2 1.2.1. Neuroanatomical abnormalities 2 1.2.2. Functional brain alterations 3 1.3. Schizophrenia aetiology 7 1.3.1. Environmental risk factors 7 1.3.2. Genetic architecture 9 1.3.2.1. Common variants 9 1.3.2.2. Rare variants 11 1.3.2.2.1. Copy number variants 11 1.3.2.2.2. Loss of function variants 12 1.3.2.2. Summary of genetic findings 14 1.4. SETD1A: utility for disease modelling 14 1.4.1. Histone modifications 14 1.4.2. H3K4 methylation 15 1.4.3. H3K4 methylation in the brain 15 1.4.4. SETD1A biochemical function 17 1.4.5. Setd1a and embryonic development 17 1.4.6. Setd1a and neurogenesis 18 1.4.7. Summary: current understanding of Setd1a function 19 1.5. Thesis aims 19 Chapter 2: General Methods 21 2.1. Animal lines 21 2.1.1. Knockout strategy 21 2.1.2. Animal husbandry 22 2.2. Genotyping 22 2.2.1. DNA extraction 22 2.2.2. PCR 22 2.2.3. Embryonic sex determination 25 2.3. Behavioural methods 25 2.3.1. Elevated plus maze 26 2.3.2. Open field test 26 v Section Page 2.3.3. Locomotor activity 26 2.3.4. Sensorimotor gating 27 2.3.5. Rotarod performance test 27 2.3.6. Novel object recognition 28 2.4. Molecular methods 29 2.4.1. Timed-matings and dissections 29 2.4.2. RNA extraction 29 2.4.3. cDNA synthesis 29 2.4.4. RT-qPCR 29 2.4.5. Protein extraction 30 2.4.6. Western blotting 30 2.5. Data analysis 31 Chapter 3: Investigating transcriptomic changes in the developing 33 brains of Setd1a+/- mice 3.1. Introduction 33 3.2. Methods 34 3.2.1. Trajectory of Setd1a expression across neurodevelopment 34 3.2.2. Generation of the model 35 3.2.3. Model validation 35 3.2.4. RT-qPCR and Western blot data analysis 36 3.2.5. RNA-seq 36 3.2.5.1. RNA extraction and quality control 36 3.2.5.2. Library preparations and sequencing 36 3.2.5.3. Analysis pipeline 36 3.2.5.4. Gene ontology enrichment analysis 38 3.2.5.5. Schizophrenia common variant gene set enrichment 38 analysis 3.3. Results 38 3.3.1. Trajectory of Setd1a expression across neurodevelopment 38 3.3.2. Model validation 39 3.3.3. RNA-seq 40 3.3.3.1. Differential expression 40 3.3.3.2. Gene ontology enrichment analysis 40 vi Section Page 3.3.3.3. Schizophrenia common variant gene set enrichment 40 analysis 3.4. Discussion 43 Chapter 4: Gestational compromise in Setd1a+/- mice and 47 consequences for growth in the early post-weaning period 4.1. Introduction 47 4.2. Methods 51 4.2.1. Timed-matings, dissections, and assessment of embryonic and 51 placental weights 4.2.2. Placental gene expression RT-qPCR 51 4.2.3. Postnatal growth 52 4.2.4. Data analysis 53 4.3. Results 53 4.3.1. E13.5 foetal and placental weight 53 4.3.2. Placental gene expression 54 4.3.3. General viability 57 4.3.4. Postnatal growth curves 57 4.4. Discussion 59 Chapter 5: Behavioural consequences of Setd1a haploinsufficiency in 63 a constitutive knockout mouse 5.1. Introduction 63 5.1.1. Aims 63 5.1.2. Anxiety 63 5.1.3. Locomotor activity 64 5.1.4. Motoric function abnormalities 65 5.1.5. Sensorimotor gating 66 5.1.6. Learning and memory 67 5.1.7. Summary 67 5.2. Methods 68 5.2.1. Behavioural testing 68 5.2.2. Data analysis 68 5.3. Results 69 5.3.1. Elevated plus maze 69 5.3.2. Open field test 70 vii Section Page 5.3.3. Locomotor activity 71 5.3.4. Sensorimotor gating 72 5.3.5. Rotarod performance test 74 5.3.6. Novel object recognition 76 5.4. Discussion 77 Chapter 6: Pharmacological modulation of sensorimotor gating 81 deficits in Setd1a+/- mice using antipsychotics 6.1. Introduction 81 6.2. Methods 84 6.2.1. Subjects 84 6.2.1.1. Pilot work 84 6.2.1.2. Main experiment 84 6.2.2. Apparatus and materials 84 6.2.2.1. Testing apparatus 84 6.2.2.2. Drugs 85 6.2.3. Design 85 6.2.3.1. Test protocol 85 6.2.3.2. Testing procedure 85 6.2.4. Data analysis 86 6.3. Results 87 6.3.1. Pilot work 87 6.3.1.1. Pilot study 1 87 6.3.1.2. Pilot study 2 88 6.3.2. Main experiment 90 6.3.2.1. Pre-test session 90 6.3.2.2. Drug challenge 91 6.3.2.2.1. Haloperidol 91 6.3.2.2.2. Risperidone 93 6.4. Discussion 95 Chapter 7: Isolating the contribution of Setd1a haploinsufficiency in the nervous system to behavioural phenotypes using a conditional 99 Setd1a KO mouse model 7.1. Introduction 99 7.2. Methods 100 viii Section Page 7.2.1. Generation of the model 100 7.2.2. Animals 100 7.2.3. Data analysis 101 7.3. Results 102 7.3.1. Model validation 102 7.3.2. Elevated plus maze 103 7.3.3. Open field test 104 7.3.4. Locomotor activity 106 7.3.5. Sensorimotor gating 108 7.3.6. Rotarod performance test 109 7.3.7. Novel object recognition 112 7.4. Discussion 113 Chapter 8: General Discussion 117 8.1. Overview 117 8.2. Setd1a haploinsufficiency causes neuropsychiatric endophenotypes 117 that cannot be rescued by antipsychotic treatments 8.3. Potential mechanisms underlying pathogenic effects of Setd1a 121 haploinsufficiency 8.3.1. Disrupted transcriptome of the developing brain 121 8.3.2. Prenatal programming by the placenta: preliminary evidence for 122 sexually dimorphic effects of Setd1a haploinsufficiency 8.3.3. Nervous system specific mechanisms 123 8.3.4. Summary 125 8.4. Limitations 126 8.4. Future directions 127 8.5. Concluding remarks 128 Bibliography 129 Appendix 1. Differentially expressed genes (Benjamini Hochberg padj < .05) 179 in E13.5 Setd1a+/- brain ix x List of Figures Figure Page Figure 1.1.
Recommended publications
  • Analyses of Allele-Specific Gene Expression in Highly Divergent
    ARTICLES Analyses of allele-specific gene expression in highly divergent mouse crosses identifies pervasive allelic imbalance James J Crowley1,10, Vasyl Zhabotynsky1,10, Wei Sun1,2,10, Shunping Huang3, Isa Kemal Pakatci3, Yunjung Kim1, Jeremy R Wang3, Andrew P Morgan1,4,5, John D Calaway1,4,5, David L Aylor1,9, Zaining Yun1, Timothy A Bell1,4,5, Ryan J Buus1,4,5, Mark E Calaway1,4,5, John P Didion1,4,5, Terry J Gooch1,4,5, Stephanie D Hansen1,4,5, Nashiya N Robinson1,4,5, Ginger D Shaw1,4,5, Jason S Spence1, Corey R Quackenbush1, Cordelia J Barrick1, Randal J Nonneman1, Kyungsu Kim2, James Xenakis2, Yuying Xie1, William Valdar1,4, Alan B Lenarcic1, Wei Wang3,9, Catherine E Welsh3, Chen-Ping Fu3, Zhaojun Zhang3, James Holt3, Zhishan Guo3, David W Threadgill6, Lisa M Tarantino7, Darla R Miller1,4,5, Fei Zou2,11, Leonard McMillan3,11, Patrick F Sullivan1,5,7,8,11 & Fernando Pardo-Manuel de Villena1,4,5,11 Complex human traits are influenced by variation in regulatory DNA through mechanisms that are not fully understood. Because regulatory elements are conserved between humans and mice, a thorough annotation of cis regulatory variants in mice could aid in further characterizing these mechanisms. Here we provide a detailed portrait of mouse gene expression across multiple tissues in a three-way diallel. Greater than 80% of mouse genes have cis regulatory variation. Effects from these variants influence complex traits and usually extend to the human ortholog. Further, we estimate that at least one in every thousand SNPs creates a cis regulatory effect.
    [Show full text]
  • Epigenetic Silencing of TGFBI Confers Resistance to Trastuzumab in Human Breast Cancer
    Palomeras et al. Breast Cancer Research (2019) 21:79 https://doi.org/10.1186/s13058-019-1160-x RESEARCH ARTICLE Open Access Epigenetic silencing of TGFBI confers resistance to trastuzumab in human breast cancer Sònia Palomeras1†, Ángel Diaz-Lagares2,3†, Gemma Viñas1,4,5, Fernando Setien2, Humberto J. Ferreira2, Glòria Oliveras1,6, Ana B. Crujeiras7,8, Alejandro Hernández4, David H. Lum9, Alana L. Welm9, Manel Esteller2,10,11,12,13* and Teresa Puig1* Abstract Background: Acquired resistance to trastuzumab is a major clinical problem in the treatment of HER2-positive (HER2+) breast cancer patients. The selection of trastuzumab-resistant patients is a great challenge of precision oncology. The aim of this study was to identify novel epigenetic biomarkers associated to trastuzumab resistance in HER2+ BC patients. Methods: We performed a genome-wide DNA methylation (450K array) and a transcriptomic analysis (RNA-Seq) comparing trastuzumab-sensitive (SK) and trastuzumab-resistant (SKTR) HER2+ human breast cancer cell models. The methylation and expression levels of candidate genes were validated by bisulfite pyrosequencing and qRT-PCR, respectively. Functional assays were conducted in the SK and SKTR models by gene silencing and overexpression. Methylation analysis in 24 HER2+ human BC samples with complete response or non-response to trastuzumab- based treatment was conducted by bisulfite pyrosequencing. Results: Epigenomic and transcriptomic analysis revealed the consistent hypermethylation and downregulation of TGFBI, CXCL2, and SLC38A1 genes in association with trastuzumab resistance. The DNA methylation and expression levels of these genes were validated in both sensitive and resistant models analyzed. Of the genes, TGFBI presented the highest hypermethylation-associated silencing both at the transcriptional and protein level.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Familial Cortical Myoclonus Caused by Mutation in NOL3 by Jonathan Foster Rnsseil DISSERTATION Submitted in Partial Satisfaction
    Familial Cortical Myoclonus Caused by Mutation in NOL3 by Jonathan Foster Rnsseil DISSERTATION Submitted in partial satisfaction of the requirements for the degree of DOCTOR OF PHILOSOPHY in Biomedical Sciences in the Copyright 2013 by Jonathan Foster Russell ii I dedicate this dissertation to Mom and Dad, for their adamantine love and support iii No man has earned the right to intellectual ambition until he has learned to lay his course by a star which he has never seen—to dig by the divining rod for springs which he may never reach. In saying this, I point to that which will make your study heroic. For I say to you in all sadness of conviction, that to think great thoughts you must be heroes as well as idealists. Only when you have worked alone – when you have felt around you a black gulf of solitude more isolating than that which surrounds the dying man, and in hope and in despair have trusted to your own unshaken will – then only will you have achieved. Thus only can you gain the secret isolated joy of the thinker, who knows that, a hundred years after he is dead and forgotten, men who never heard of him will be moving to the measure of his thought—the subtile rapture of a postponed power, which the world knows not because it has no external trappings, but which to his prophetic vision is more real than that which commands an army. -Oliver Wendell Holmes, Jr. iv ACKNOWLEDGMENTS I am humbled by the efforts of many, many others who were essential for this work.
    [Show full text]
  • Transcriptome-Wide Profiling of the Neonatal Monocyte Response to E
    Transcriptome-wide profiling of the neonatal monocyte response to E. coli and S. epidermidis Emma de Jong, B.Sc. (Hons) This thesis is presented for the degree of Doctor of Philosophy School of Veterinary & Life Sciences 2015 Declaration I declare that this thesis is my own account of my research, and contains as its main content work that has not previously been submitted for a degree at any other tertiary education institution. .................................... (Emma de Jong) i ii Abstract Preterm infants are extremely vulnerable to life-threatening invasive infections (particularly with Staphylococcus epidermidis and Escherichia coli) however our understanding of their innate immune defences is limited. Furthermore, prenatal exposure to histologic chorioamnionitis (HCA) complicates 40–70% of preterm births and is known to modulate the risk for sepsis, yet the impact of HCA on the development of innate immunity is largely unknown. We hypothesised that inadequate monocyte activation by neonatal pathogens results in impaired innate immune responses thereby increasing preterm infants’ susceptibility to invasive infection, and that prenatal exposure to HCA overrides these impairments. Cell sorting and bacterial stimulation methodologies were developed and optimised specifically for working with human infant cord blood samples. RNA-sequencing was performed on purified cord blood monocytes from very preterm (≤31 weeks gestational age (GA)) and term infants (37–40 weeks GA) following challenge with live S. epidermidis or E. coli to identify gene/pathway differences specific to the preterm infant. Protein levels of inflammatory cytokines and chemokines were measured in paired monocyte culture supernatants. Preterm infants displayed a quantitative monocyte deficiency compared to term infants, manifesting as reduced frequencies of classical monocytes with significantly reduced CD14 expression.
    [Show full text]
  • Termination of RNA Polymerase II Transcription by the 5’-3’ Exonuclease Xrn2
    TERMINATION OF RNA POLYMERASE II TRANSCRIPTION BY THE 5’-3’ EXONUCLEASE XRN2 by MICHAEL ANDRES CORTAZAR OSORIO B.S., Universidad del Valle – Colombia, 2011 A thesis submitted to the Faculty of the Graduate School of the University of Colorado in partial fulfillment of the requirements for the degree of Doctor of Philosophy Molecular Biology Program 2018 This thesis for the Doctor of Philosophy degree by Michael Andrés Cortázar Osorio has been approved for the Molecular Biology Program by Mair Churchill, Chair Richard Davis Jay Hesselberth Thomas Blumenthal James Goodrich David Bentley, Advisor Date: Aug 17, 2018 ii Cortázar Osorio, Michael Andrés (Ph.D., Molecular Biology) Termination of RNA polymerase II transcription by the 5’-3’ exonuclease Xrn2 Thesis directed by Professor David L. Bentley ABSTRACT Termination of transcription occurs when RNA polymerase (pol) II dissociates from the DNA template and releases a newly-made mRNA molecule. Interestingly, an active debate fueled by conflicting reports over the last three decades is still open on which of the two main models of termination of RNA polymerase II transcription does in fact operate at 3’ ends of genes. The torpedo model indicates that the 5’-3’ exonuclease Xrn2 targets the nascent transcript for degradation after cleavage at the polyA site and chases pol II for termination. In contrast, the allosteric model asserts that transcription through the polyA signal induces a conformational change of the elongation complex and converts it into a termination-competent complex. In this thesis, I propose a unified allosteric-torpedo mechanism. Consistent with a polyA site-dependent conformational change of the elongation complex, I found that pol II transitions at the polyA site into a mode of slow transcription elongation that is accompanied by loss of Spt5 phosphorylation in the elongation complex.
    [Show full text]
  • Identification of Protein Partners for Nibp, a Novel Nik-And Ikkb-Binding Protein Through Experimental, Computational and Bioinformatics Techniques
    IDENTIFICATION OF PROTEIN PARTNERS FOR NIBP, A NOVEL NIK-AND IKKB-BINDING PROTEIN THROUGH EXPERIMENTAL, COMPUTATIONAL AND BIOINFORMATICS TECHNIQUES A Thesis Submitted to the Temple University Graduate Board In Partial Fulfillment of the Requirements for the Degree MASTER OF SCIENCE by Sombudha Adhikari August 2013 Thesis Approvals: Mohammad. F. Kiani, Ph.D., Thesis Advisor, College of Engineering Wenhui Hu, M.D., Ph.D., School of Medicine Roland Dunbrack, Ph.D, Fox Chase Cancer Center Peter Lelkes, Ph.D., College of Engineering ABSTRACT Identification of protein partners for NIBP, a novel NIK- and IKK β-binding protein through experimental, computational and bioinformatics techniques NIBP is a prototype member of a novel protein family. It forms a novel subcomplex of NIK-NIBP-IKK β and enhances cytokine-induced IKK β-mediated NF κB activation. It is also named TRAPPC9 as a key member of trafficking particle protein (TRAPP) complex II, which is essential in trans -Golgi networking (TGN). The signaling pathways and molecular mechanisms for NIBP actions remain largely unknown. The aim of this research is to identify potential proteins interacting with NIBP, resulting in the regulation of NF κB signaling pathways and other unknown signaling pathways. At Dr. Wenhui Hu’s lab in the Department of Neuroscience, Temple University, sixteen partner proteins were experimentally identified that potentially bind to NIBP. NIBP is a novel protein with no entry in the Protein Data Bank. From a computational and bioinformatics standpoint, we use prediction of secondary structure and protein disorder as well as homology-based structural modeling approaches to create a hypothesis on protein-protein interaction between NIBP and the partner proteins.
    [Show full text]
  • Exploration of the Relationships Between Tumor Mutation Burden with Immune Infiltrates in Clear Cell Renal Cell Carcinoma
    648 Original Article Page 1 of 16 Exploration of the relationships between tumor mutation burden with immune infiltrates in clear cell renal cell carcinoma Chuanjie Zhang1#, Zongtai Li2#, Feng Qi3#, Xin Hu4, Jun Luo5 1Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; 2Department of Medical Oncology, Gaozhou People’s Hospital, Gaozhou 525200, China; 3Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; 4First Clinical Medical College of Nanjing Medical University, Nanjing 210029, China; 5Department of Urology, Shanghai Fourth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai 200081, China Contributions: (I) Conception and design: J Luo, C Zhang; (II) Administrative support: C Zhang; (III) Provision of study materials or patients: Z Li, F Qi; (IV) Collection and assembly of data: X Hu; (V) Data analysis and interpretation: J Luo, C Zhang; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors. #These authors contributed equally to this article. Correspondence to: Jun Luo. Department of Urology, Shanghai Fourth People’s Hospital affiliated to Tongji University School of Medicine, No. 1878 North Sichuan Road, Hongkou District, Shanghai 200081, China. Email: [email protected]. Background: Whether tumor mutation burden (TMB) correlated with improved survival outcomes or promotion of immunotherapies remained controversy in various malignancies. We aimed to investigate the prognosis of TMB and the potential association with immune infiltrates in clear cell renal cell carcinoma (ccRCC). Methods: We downloaded the somatic mutation data of 336 ccRCC patients from the Cancer Genome Atlas (TCGA) database, and analyzed the mutation profiles with “maftools” package.
    [Show full text]
  • Genome-Wide Meta-Analysis Identifies Six Novel Loci Associated With
    Molecular Psychiatry (2015) 20, 647–656 © 2015 Macmillan Publishers Limited All rights reserved 1359-4184/15 www.nature.com/mp ORIGINAL ARTICLE Genome-wide meta-analysis identifies six novel loci associated with habitual coffee consumption The Coffee and Caffeine Genetics Consortium, MC Cornelis1,2, EM Byrne3,117, T Esko4,5,6,7,117, MA Nalls8,117, A Ganna9, N Paynter10, KL Monda11, N Amin12, K Fischer4, F Renstrom13, JS Ngwa14, V Huikari15, A Cavadino16, IM Nolte17, A Teumer18,KYu19, P Marques-Vidal20, R Rawal21, A Manichaikul22, MK Wojczynski23, JM Vink24, JH Zhao25, G Burlutsky26, J Lahti27,28, V Mikkilä29,30, RN Lemaitre31, J Eriksson32, SK Musani33, T Tanaka34, F Geller35, J Luan25, J Hui36,37,38,39, R Mägi4, M Dimitriou40, ME Garcia41, W-K Ho42, MJ Wright43, LM Rose10, PKE Magnusson9, NL Pedersen9, D Couper44, BA Oostra45, A Hofman12, MA Ikram12,46,47, HW Tiemeier12,48, AG Uitterlinden12,49, FJA van Rooij12, I Barroso50,51, I Johansson52, L Xue14, M Kaakinen15,53,54, L Milani4, C Power16, H Snieder17, RP Stolk17, SE Baumeister55, R Biffar56,FGu19, F Bastardot57, Z Kutalik58,59,60, DR Jacobs Jr61, NG Forouhi25, E Mihailov4, L Lind62, C Lindgren63, K Michaëlsson64, A Morris63, M Jensen2, K-T Khaw42, RN Luben42, JJ Wang26, S Männistö65, M-M Perälä65, M Kähönen66, T Lehtimäki67, J Viikari68, D Mozaffarian1,2,69,70, K Mukamal71, BM Psaty31,72,73,74, A Döring75, AC Heath76, GW Montgomery43, N Dahmen77, T Carithers78, KL Tucker79, L Ferrucci34, HA Boyd35, M Melbye35, JL Treur24, D Mellström32, JJ Hottenga24, I Prokopenko63,80, A Tönjes81,82,
    [Show full text]
  • Downloaded Per Proteome Cohort Via the Web- Site Links of Table 1, Also Providing Information on the Deposited Spectral Datasets
    www.nature.com/scientificreports OPEN Assessment of a complete and classifed platelet proteome from genome‑wide transcripts of human platelets and megakaryocytes covering platelet functions Jingnan Huang1,2*, Frauke Swieringa1,2,9, Fiorella A. Solari2,9, Isabella Provenzale1, Luigi Grassi3, Ilaria De Simone1, Constance C. F. M. J. Baaten1,4, Rachel Cavill5, Albert Sickmann2,6,7,9, Mattia Frontini3,8,9 & Johan W. M. Heemskerk1,9* Novel platelet and megakaryocyte transcriptome analysis allows prediction of the full or theoretical proteome of a representative human platelet. Here, we integrated the established platelet proteomes from six cohorts of healthy subjects, encompassing 5.2 k proteins, with two novel genome‑wide transcriptomes (57.8 k mRNAs). For 14.8 k protein‑coding transcripts, we assigned the proteins to 21 UniProt‑based classes, based on their preferential intracellular localization and presumed function. This classifed transcriptome‑proteome profle of platelets revealed: (i) Absence of 37.2 k genome‑ wide transcripts. (ii) High quantitative similarity of platelet and megakaryocyte transcriptomes (R = 0.75) for 14.8 k protein‑coding genes, but not for 3.8 k RNA genes or 1.9 k pseudogenes (R = 0.43–0.54), suggesting redistribution of mRNAs upon platelet shedding from megakaryocytes. (iii) Copy numbers of 3.5 k proteins that were restricted in size by the corresponding transcript levels (iv) Near complete coverage of identifed proteins in the relevant transcriptome (log2fpkm > 0.20) except for plasma‑derived secretory proteins, pointing to adhesion and uptake of such proteins. (v) Underrepresentation in the identifed proteome of nuclear‑related, membrane and signaling proteins, as well proteins with low‑level transcripts.
    [Show full text]
  • Analysis of a Putative Imprinted Locus Within the TRAPPC9 Intellectual Disability Gene
    Analysis of a putative imprinted locus within the TRAPPC9 intellectual disability gene by Rosalind Law A thesis submitted in conformity with the requirement for the degree of Master of Science Institute of Medical Science University of Toronto © by Rosalind Law 2014 ii Analysis of a putative imprinted locus within the TRAPPC9 intellectual disability gene Rosalind Law Master of Science Institute of Medical Science University of Toronto 2014 Individuals were identified carrying heterozygous copy number variations within the autosomal recessive intellectual disability gene TRAPPC9 (chr8q24.3) and lacking apparent mutations on the second allele, thus indicating the involvement of a conserved imprinting mechanism at this locus between mice (15qD3) and humans. My primary objective was to identify whether a potentially equivalent to mouse Peg13, PEG13, located within an intron of TRAPPC9, was imprinted in various human tissues and cell lines. To investigate imprinting at this locus, allelic expression of PEG13 was assessed using Sanger sequencing and pyrosequencing. To identify regulatory regions that may be responsible for variations in PEG13 allelic expression, methylation profiles at CpG islands were determined in human tissues and cell lines. The PEG13 equivalent was found preferentially-expressed from the paternal allele. Its imprinted expression appears to be tissue-specific and conserved in human fetal brain. Expression patterns of PEG13 in human tissues of paralleled KCNK9. iii Acknowledgements I feel incredibly grateful for the opportunities I have experienced and the people I have encountered in the past two years. I am also thankful for those who have guided me through the unpredictable, yet rewarding field of medical research.
    [Show full text]
  • TRAPPC9-Related Autosomal Recessive Intellectual Disability: Report of a New Mutation and Clinical Phenotype
    European Journal of Human Genetics (2013) 21, 229–232 & 2013 Macmillan Publishers Limited All rights reserved 1018-4813/13 www.nature.com/ejhg SHORT REPORT TRAPPC9-related autosomal recessive intellectual disability: report of a new mutation and clinical phenotype Giuseppe Marangi1, Vincenzo Leuzzi2, Filippo Manti2, Serena Lattante1, Daniela Orteschi1, Vanna Pecile3, Giovanni Neri1 and Marcella Zollino*,1 Intellectual disability (ID) with autosomal recessive (AR) inheritance is believed to be common; however, very little is known about causative genes and genotype–phenotype correlations. The broad genetic heterogeneity of AR-ID, and its usually nonsyndromic nature make it difficult to pool multiple pedigrees with the same underlying genetic defect to achieve consistent nosology. Nearly all autosomal genes responsible for recessive cognitive disorders have been identified in large consanguineous families from the Middle East, and nonsense mutations in TRAPPC9 have been reported in a total of 5. Although several recurrent phenotypic abnormalities are described in some of these patients, the associated phenotype is usually referred to as nonsyndromic. By means of single-nucleotide polymorphism-array first and then by exome sequencing, we identified a new pathogenic mutation in TRAPPC9 in two Italian sisters born to healthy and apparently nonconsanguineous parents. It consists of a homozygous splice site mutation causing exon skipping with frameshift and premature termination, as confirmed by mRNA sequencing. By detailed phenotypic analysis of our patients, and by critical literature review, we found that homozygous TRAPPC9 loss-of-function mutations cause a distinctive phenotype, characterized by peculiar facial appearance, obesity, hypotonia (all signs resembling a Prader–Willi-like phenotype), moderate-to-severe ID, and consistent brain abnormalities.
    [Show full text]