Supplemental Figure 1

Total Page:16

File Type:pdf, Size:1020Kb

Supplemental Figure 1 Supplemental Figure 1 la avb san ra avn lv Tbx3 Tbx5 Scn5a Supplemental Figure 1 Cartoon depicting expression patterns of Tbx3, Tbx5 and Scn5a in the heart from fetal stages onwards. Tbx5 activates Scn5a in the heart. The pattern of Scn5a is broader than that of Tbx5, indicating other activators expressed in broader patterns are involved in Scn5a activation. Note the absence of expression of Scn5a in the Tbx3+ (red) and Tbx5+ (purple) region of the sinus node (san), internodal region, atrioventricular node (avn) and atrioventricular canal myocardium, indicating that the repressive function of Tbx3 is dominant in these parts of the heart. avb, atrioventricular bundle; l/ra, left/right atrium; l/rv, left/right ventricle. Supplemental Figure 2 20 kbp 30 Tbx3 3 25 Gata4 3 75 5 - Nkx2 9 50 p300 0 Tbx5* Hopx Gata4* Nkx2-5* P300# P300$ Pol2$ Supplemental Figure 2 Alignment of ChIP-seq datasets from mouse heart (Tbx3, red; Gata4, blue; Nkx2-5, green; p300, orange) described in this manuscript, with other published datasets in the vicinity of Hopx, a gene involved in conduction system function (1). Overlap in multiple datasets was used as selection criterion for enhancer activity screening (Figure 1C). Supplemental Figure 3 Tbx5 TRANSFAC GATA1 TRANSFAC NKX2-5 JASPAR T JASPAR GATA1 JASPAR NKX3-2 JASPAR HL1 Tbx5 ChIP-seq motif HL1 Gata4 ChIP-seq motif HL1 Nkx2-5 ChIP-seq motif TBX3 ChIP-seq motif Gata4 ChIP-seq motif Nkx2-5 ChIP-seq motif Supplemental Figure 3 Motif discovery analysis for ChIP-seq data. MEME motif discovery analysis for Tbx3 ChIP-seq data, Gata4 ChIP- seq data and Nkx2-5 ChIP-seq data (bottom row) compared to published consensus sequences available from TRANSFAC (2), JASPAR (3,4) and the recently published HL-1 ChIP-seq derived binding sites (5). Of note is the closer resemblance of the ChIP-seq experimentally derived Nkx2-5 sites to the NKX3-2 JASPAR consensus site than that of the NKX2-5 JASPAR consensus site. Supplemental Figure 4 A GOTERM: Biological Process Genes Count % P-value Cytoskeleton organization 8 8.9 1.4E-4 Actin cytoskeleton organization 5 5.6 1.0E-2 Actin filament-based process 5 5.6 1.3E-2 Heart development 5 5.6 2.8E-2 Actin filament organization 3 3.3 3.4E-2 Muscle contraction 3 3.3 3.8E-2 Intracellular signalling cascade 3 3.3 4.4E-2 Muscle system process 10 11.1 4.7E-2 Σn genes with Tbx3 peak Significance of B Σn genes (% of Σn genes) enrichment (Z test) Enriched in AVC 2580 1329 (51.5) p=6.7x10-10 Reduced in AVC 2627 1635 (62.2) p=3.9x10-43 non-enriched controls (N=6) 2545 x 1022 (40.2±2.5) Tbx3 induced 578 354 (61.2) p=2.3x10-12 Tbx3 reduced 524 347 (66.2) p=2.8x10-17 non-enriched controls (N=6) 525 197. 5(37.6±2.6) Supplemental Figure 4 Overlap between ChIP-seq datasets and expression arrays. (A) Molecular function GO-term analysis of genes containing an intragenic overlapping Tbx3/Nkx2-5/Gata4-ChIP-seq peak region. (B) Overlap between Tbx3 ChIP-seq dataset and Tbx3 microarray datasets. Intersection of Tbx3 ChIP-seq peaks, with genes found significantly (P < 0.05) enriched or reduced in the AVC myocardium, in the induced TBX3 expression system or in the control gene microarray sets. There are significantly more intersecting genes found in the reduced AVC array set compared the induced set (P < 0.001). The same holds for the reduced set of genes in the induced TBX3 expression model compared to the induced set of genes (P < 0.05). Supplemental Figure 5 Tbx3 embryonic heart 1000 Tbx5 adult heart 100 Hprt over over 10 Neg Enrichment 1 Krr1 Hprt Sbds Otos Eif3d Cog5 Gbp4 Sept9 Myh6 Chsy1 Grh13 Iqsec1 Isoc2a Frmd6 Tcmo7 Rbpms Wdr95 Ccrn41 Bcl2l11 Irf2bp2 Chchd6 Ppap2b Frmd4a Slc22a2 Gabarap Tead1 F2 0 Intergenic Supplemental Figure 5 ChIP-qPCR showing relative enrichment of randomly chosen ChIP-seq peaks over a negative control region in the Hprt gene for Tbx3 in embryonic heart (ED10.5, pink) and Tbx5 in adult heart (purple). Supplemental Figure 6 250% 200% ] 150% p < 0.05 mRNA [ control 100% Induced Tbx3 Relative 50% * 0% Gapdh Hprt Nkx2-5 Gata4 Gja1 Supplemental Figure 6 Quantitative PCR showing that housekeeping genes Gapdh and Hprt, used for normalization of the other genes, are not affected by the presence of Tbx3. Also transcription factors Nkx2-5 and Gata4, working in complex with Tbx3, are unaffected by the presence of Tbx3, while Gja1 (Cx43), a known target of Tbx3, is significantly down regulated by Tbx3. Error bars represent standard deviation. Supplemental Figure 7 Scn5a Scn10a A 0,2 0,4 Limb Brain 0,2 0,1 Atria Ventricle relative [mRNA] relative relative [mRNA] relative 0,0 0,0 E10.5 E14.5 E17.5 Adult E10.5 E14.5 E17.5 Adult Liver B 1,4 C 1,2 Input ChIP Target ] 1 PCR co Tbx3 co Tbx3 0,8 4 mRNA [ * 0,6 Control 9 0,4 a relative * Induced TBX3 0,2 b 0 Hprt Supplemental Figure 7 Expression and ChIP analysis of Scn5a / Scn10a locus (A) Relative mRNA expression levels of Scn5a and Scn10a in limb, brain, atria, ventricles and liver of E10.5, E14.5, E17.5 and adult mice. Both genes are primarily expressed in atria and ventricles. (B) qRT-PCR analysis shows down regulation of Scn5a and Scn10a in atria of induced TBX3 expressing mice (black bars) as compared to control mice (light grey bars). Both genes are reduced in the AV canal with a p-value < 0.05 and contain ChIP-seq peaks for Tbx3, Gata4 and Nkx2-5. Flanking genes Scn11a and Exog are unaffected by induced TBX3 expression and do not contain peaks for Nkx2-5, Gata4 and Tbx3. (C) ChIP-PCR on hearts of adult induced TBX3 expressing mice compared to control mice. Figure shows an enrichment of fragment 4 and 9, while there is no enrichment for control regions a and b (Figure 4A). Supplemental Figure 8 A F9 B 2 2 2 Tbx3 F1 * F2 F9 * 1 Gata4 1 1 RLU * RLU * RLU 5 * - * * * 0 0 0 Nkx2 - Tbx5 Tbx3 Tbx2 - Tbx2 Tbx5 Tbx3 Tbx3 - Tbx5 Tbx2 p300 C minP F9-A F9-B F9-C F9-D F9-E F9-F F9-G - F9-H Nkx2-5 + Gata4 F9-I Nkx2-5 + Gata4 + VP16-TBX2 RLU 0 50 100 150 200 250 Supplemental Figure 8 Transfection assays with Tbox factors on enhancer candidates. (A) Close-up of the alignment of ChIP-seq datasets Tbx3 (red), Gata4 (blue), Nkx2-5 (green) and p300 (orange) for fragment 9. (B) F1 and 2 are activated by Tbx5 and repressed by Tbx2 and Tbx3. F9 was not stimulated by Tbx5, but responds to repression with Tbx2 and Tbx3. (C) Shortening of 5' flanking sequences in fragment 9 results in an increased response to Gata4, Nkx2-5 and Tbx2-VP16 (activating fusion protein) and identifies a small, responsive element that harbors binding sites for all TFs. Experiments were performed in Cos7 cells. Supplemental Figure 9 1.2 p<0.001 1.0 p < 0.001 0.8 0.6 * 0.4 * * * 0.2 0.0 B Maj1 Min1 Mut Supplemental Figure 9 Quantitative analysis of EMSA data. Non-radioactive EMSA was performed 3 times and after chemilumiscent imaging (see Figure 6B for representative image), the results were quantified using AIDA software. Under these conditions, the allele containing SNP rs6801957 (Min1) shows a significant decrease of approximately 60% in T-box binding compared to the major allele (Maj1). B is average background signal, Mut shows signal of a probe in which the three base pairs of the T-box binding site were mutated. REFERENCES (1) Ismat FA et al. Homeobox protein Hop functions in the adult cardiac conduction system. Circ Res. 2005;96(8):898-903. (2) Matys V et al. TRANSFAC: transcriptional regulation, from patterns to profiles. Nucleic Acids Res. 2003;31(1):374-378. (3) Stormo GD. DNA binding sites: representation and discovery. Bioinformatics. 2000;16(1):16-23. (4) Wasserman WW, Sandelin A. Applied bioinformatics for the identification of regulatory elements. Nat Rev Genet. 2004;5(4):276-287. (5) He A, Kong SW, Ma Q, Pu WT. Co-occupancy by multiple cardiac transcription factors identifies transcriptional enhancers active in heart. Proc Natl Acad Sci U S A. 2011;108:5632-5637. Supplemental Methods ChIP-peak calling For peak calling, a minimum binding site length was set to 15 bp and regions in closer proximity than 50 bp were concatenated into one region. For identification of regions with robust binding, a transcription factor-dependent (see below) minimum number of reads per peak was set. The rationale behind the transcription factor-dependent thresholds was the difference in signal to noise ratios between different transcription factors. The thresholds were calculated as follows: The log of the number of peaks called at different thresholds (y- axis) was plotted against the threshold used (x-axis). This plot was typically biphasic, reflecting a transition from random to genuine binding events at the point where the second derivative is maximal. For the Tbx3 data set, for example, this phase change was found at a tag-count of 5 (maximum of the second derivative) and for Nkx2-5 at a count of 15. To identify the most robust binding sites used in the analyses throughout the paper, a threshold of two times the number of reads found at the maximum of the second derivative was applied.
Recommended publications
  • Table S1. List of Proteins in the BAHD1 Interactome
    Table S1. List of proteins in the BAHD1 interactome BAHD1 nuclear partners found in this work yeast two-hybrid screen Name Description Function Reference (a) Chromatin adapters HP1α (CBX5) chromobox homolog 5 (HP1 alpha) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins (20-23) HP1β (CBX1) chromobox homolog 1 (HP1 beta) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins HP1γ (CBX3) chromobox homolog 3 (HP1 gamma) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins MBD1 methyl-CpG binding domain protein 1 Binds methylated CpG dinucleotide and chromatin-associated proteins (22, 24-26) Chromatin modification enzymes CHD1 chromodomain helicase DNA binding protein 1 ATP-dependent chromatin remodeling activity (27-28) HDAC5 histone deacetylase 5 Histone deacetylase activity (23,29,30) SETDB1 (ESET;KMT1E) SET domain, bifurcated 1 Histone-lysine N-methyltransferase activity (31-34) Transcription factors GTF3C2 general transcription factor IIIC, polypeptide 2, beta 110kDa Required for RNA polymerase III-mediated transcription HEYL (Hey3) hairy/enhancer-of-split related with YRPW motif-like DNA-binding transcription factor with basic helix-loop-helix domain (35) KLF10 (TIEG1) Kruppel-like factor 10 DNA-binding transcription factor with C2H2 zinc finger domain (36) NR2F1 (COUP-TFI) nuclear receptor subfamily 2, group F, member 1 DNA-binding transcription factor with C4 type zinc finger domain (ligand-regulated) (36) PEG3 paternally expressed 3 DNA-binding transcription factor with
    [Show full text]
  • Update on the Implication of Potassium Channels in Autism: K+ Channelautism Spectrum Disorder
    REVIEW ARTICLE published: 02 March 2015 doi: 10.3389/fncel.2015.00034 Update on the implication of potassium channels in autism: K+ channelautism spectrum disorder Luca Guglielmi 1*, Ilenio Servettini 1, Martino Caramia 2 , Luigi Catacuzzeno 2 , Fabio Franciolini 2 , Maria Cristina D’Adamo1 and Mauro Pessia1* 1 Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy 2 Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy Edited by: Autism spectrum disorders (ASDs) are characterized by impaired ability to properly Tycho M. Hoogland, Netherlands implement environmental stimuli that are essential to achieve a state of social and Institute for Neuroscience, Netherlands cultural exchange. Indeed, the main features of ASD are impairments of interpersonal relationships, verbal and non-verbal communication and restricted and repetitive behaviors. Reviewed by: Noritaka Ichinohe, National Institute of These aspects are often accompanied by several comorbidities such as motor delay, Neuroscience – National Center of praxis impairment, gait abnormalities, insomnia, and above all epilepsy. Genetic analyses Neurology and Psychiatry, Japan of autistic individuals uncovered deleterious mutations in several K+ channel types Mohamed Jaber, University of Poitiers, France strengthening the notion that their intrinsic dysfunction may play a central etiologic role in ASD. However, indirect implication of K+ channels in ASD has been also reported. *Correspondence: + Luca Guglielmi and Mauro Pessia, For instance, loss of fragile X mental retardation protein (FMRP) results in K channels Section of Physiology and deregulation, network dysfunction and ASD-like cognitive and behavioral symptoms. This Biochemistry, Department of review provides an update on direct and indirect implications of K+ channels in ASDs.
    [Show full text]
  • An Integrative Study
    Published OnlineFirst January 12, 2010; DOI: 10.1158/1535-7163.MCT-09-0321 Molecular Spotlight on Molecular Profiling Cancer Therapeutics Multifactorial Regulation of E-Cadherin Expression: An Integrative Study William C. Reinhold1, Mark A. Reimers1,2, Philip Lorenzi1,3, Jennifer Ho1, Uma T. Shankavaram1,4, Micah S. Ziegler1, Kimberly J. Bussey1,5, Satoshi Nishizuka1,6, Ogechi Ikediobi1,7, Yves G. Pommier1, and John N. Weinstein1,3 Abstract E-cadherin (E-cad) is an adhesion molecule associated with tumor invasion and metastasis. Its down- regulation is associated with poor prognosis for many epithelial tumor types. We have profiled E-cad in the NCI-60 cancer cell lines at the DNA, RNA, and protein levels using six different microarray platforms plus bisulfite sequencing. Here we consider the effects on E-cad expression of eight potential regulatory factors: E-cad promoter DNA methylation, the transcript levels of six transcriptional repressors (SNAI1, SNAI2, TCF3, TCF8, TWIST1, and ZFHX1B), and E-cad DNA copy number. Combined bioinformatic and pharmacological analyses indicate the following ranking of influence on E-cad expression: (1) E-cad pro- moter methylation appears predominant, is strongly correlated with E-cad expression, and shows a 20% to 30% threshold above which E-cad expression is silenced; (2) TCF8 expression levels correlate with (−0.62) and predict (P < 0.00001) E-cad expression; (3) SNAI2 and ZFHX1B expression levels correlate positively with each other (+0.83) and also correlate with (−0.32 and −0.30, respectively) and predict (P =0.03and 0.01, respectively) E-cad expression; (4) TWIST1 correlates with (−0.34) but does not predict E-cad expres- sion; and (5) SNAI1 expression, TCF3 expression, and E-cad DNA copy number do not correlate with or predict E-cad expression.
    [Show full text]
  • Towards Mutation-Specific Precision Medicine in Atypical Clinical
    International Journal of Molecular Sciences Review Towards Mutation-Specific Precision Medicine in Atypical Clinical Phenotypes of Inherited Arrhythmia Syndromes Tadashi Nakajima * , Shuntaro Tamura, Masahiko Kurabayashi and Yoshiaki Kaneko Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi 371-8511, Gunma, Japan; [email protected] (S.T.); [email protected] (M.K.); [email protected] (Y.K.) * Correspondence: [email protected]; Tel.: +81-27-220-8145; Fax: +81-27-220-8158 Abstract: Most causal genes for inherited arrhythmia syndromes (IASs) encode cardiac ion channel- related proteins. Genotype-phenotype studies and functional analyses of mutant genes, using heterol- ogous expression systems and animal models, have revealed the pathophysiology of IASs and enabled, in part, the establishment of causal gene-specific precision medicine. Additionally, the utilization of induced pluripotent stem cell (iPSC) technology have provided further insights into the patho- physiology of IASs and novel promising therapeutic strategies, especially in long QT syndrome. It is now known that there are atypical clinical phenotypes of IASs associated with specific mutations that have unique electrophysiological properties, which raises a possibility of mutation-specific precision medicine. In particular, patients with Brugada syndrome harboring an SCN5A R1632C mutation exhibit exercise-induced cardiac events, which may be caused by a marked activity-dependent loss of R1632C-Nav1.5 availability due to a marked delay of recovery from inactivation. This suggests that the use of isoproterenol should be avoided. Conversely, the efficacy of β-blocker needs to be examined. Patients harboring a KCND3 V392I mutation exhibit both cardiac (early repolarization syndrome and Citation: Nakajima, T.; Tamura, S.; paroxysmal atrial fibrillation) and cerebral (epilepsy) phenotypes, which may be associated with a Kurabayashi, M.; Kaneko, Y.
    [Show full text]
  • Mediator of DNA Damage Checkpoint 1 (MDC1) Is a Novel Estrogen Receptor Co-Regulator in Invasive 6 Lobular Carcinoma of the Breast 7 8 Evelyn K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.16.423142; this version posted December 16, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. 1 Running Title: MDC1 co-regulates ER in ILC 2 3 Research article 4 5 Mediator of DNA damage checkpoint 1 (MDC1) is a novel estrogen receptor co-regulator in invasive 6 lobular carcinoma of the breast 7 8 Evelyn K. Bordeaux1+, Joseph L. Sottnik1+, Sanjana Mehrotra1, Sarah E. Ferrara2, Andrew E. Goodspeed2,3, James 9 C. Costello2,3, Matthew J. Sikora1 10 11 +EKB and JLS contributed equally to this project. 12 13 Affiliations 14 1Dept. of Pathology, University of Colorado Anschutz Medical Campus 15 2Biostatistics and Bioinformatics Shared Resource, University of Colorado Comprehensive Cancer Center 16 3Dept. of Pharmacology, University of Colorado Anschutz Medical Campus 17 18 Corresponding author 19 Matthew J. Sikora, PhD.; Mail Stop 8104, Research Complex 1 South, Room 5117, 12801 E. 17th Ave.; Aurora, 20 CO 80045. Tel: (303)724-4301; Fax: (303)724-3712; email: [email protected]. Twitter: 21 @mjsikora 22 23 Authors' contributions 24 MJS conceived of the project. MJS, EKB, and JLS designed and performed experiments. JLS developed models 25 for the project. EKB, JLS, SM, and AEG contributed to data analysis and interpretation. SEF, AEG, and JCC 26 developed and performed informatics analyses. MJS wrote the draft manuscript; all authors read and revised the 27 manuscript and have read and approved of this version of the manuscript.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • The Direction of Cross Affects Obesity After Puberty in Male but Not Female
    Kärst et al. BMC Genomics (2015) 16:904 DOI 10.1186/s12864-015-2164-2 RESEARCH ARTICLE Open Access The direction of cross affects obesity after puberty in male but not female offspring Stefan Kärst1†, Danny Arends1†, Sebastian Heise1†, Jan Trost1, Marie-Laure Yaspo2, Vyacheslav Amstislavskiy2, Thomas Risch2, Hans Lehrach2 and Gudrun A. Brockmann1* Abstract Background: We investigated parent-of-origin and allele-specific expression effects on obesity and hepatic gene expression in reciprocal crosses between the Berlin Fat Mouse Inbred line (BFMI) and C57Bl/6NCrl (B6N). Results: We found that F1-males with a BFMI mother developed 1.8 times more fat mass on a high fat diet at 10 weeks than F1-males of a BFMI father. The phenotype was detectable from six weeks on and was preserved after cross-fostering. RNA-seq data of liver provided evidence for higher biosynthesis and elongation of fatty acids (p = 0.00635) in obese male offspring of a BFMI mother versus lean offspring of a BFMI father. Furthermore, fatty acid degradation (p = 0.00198) and the peroxisome pathway were impaired (p = 0.00094). The circadian rhythm was affected as well (p = 0.00087). Among the highest up-regulated protein coding genes in obese males were Acot4 (1.82 fold, p = 0.022), Cyp4a10 (1.35 fold, p = 0.026) and Cyp4a14 (1.32 fold, p = 0.012), which hydroxylize fatty acids and which are known to be increased in liver steatosis. Obese males showed lower expression of the genetically imprinted and paternally expressed 3 (Peg3) gene (0.31 fold, p = 0.046) and higher expression of the androgen receptor (Ar) gene (2.38 fold, p = 0.068).
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • Contribution of Enhancer-Driven and Master-Regulator Genes to Autoimmune Disease Revealed Using Functionally Informed SNP-To-Gene Linking Strategies Kushal K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.09.02.279059; this version posted March 31, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Contribution of enhancer-driven and master-regulator genes to autoimmune disease revealed using functionally informed SNP-to-gene linking strategies Kushal K. Dey1, Steven Gazal1, Bryce van de Geijn 1,3, Samuel Sungil Kim 1,4, Joseph Nasser5, Jesse M. Engreitz5, Alkes L. Price 1,2,5 1 Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA 2 Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 3 Genentech, South San Francisco, CA 4 Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 5 Broad Institute of MIT and Harvard, Cambridge, MA, USA Abstract Gene regulation is known to play a fundamental role in human disease, but mechanisms of regulation vary greatly across genes. Here, we explore the con- tributions to disease of two types of genes: genes whose regulation is driven by enhancer regions as opposed to promoter regions (Enhancer-driven) and genes that regulate many other genes in trans (Master-regulator). We link these genes to SNPs using a comprehensive set of SNP-to-gene (S2G) strategies and apply stratified LD score regression to the resulting SNP annotations to draw three main conclusions about 11 autoimmune diseases and blood cell traits (average Ncase=13K across 6 autoimmune diseases, average N=443K across 5 blood cell traits).
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Accompanies CD8 T Cell Effector Function Global DNA Methylation
    Global DNA Methylation Remodeling Accompanies CD8 T Cell Effector Function Christopher D. Scharer, Benjamin G. Barwick, Benjamin A. Youngblood, Rafi Ahmed and Jeremy M. Boss This information is current as of October 1, 2021. J Immunol 2013; 191:3419-3429; Prepublished online 16 August 2013; doi: 10.4049/jimmunol.1301395 http://www.jimmunol.org/content/191/6/3419 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2013/08/20/jimmunol.130139 Material 5.DC1 References This article cites 81 articles, 25 of which you can access for free at: http://www.jimmunol.org/content/191/6/3419.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on October 1, 2021 • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Global DNA Methylation Remodeling Accompanies CD8 T Cell Effector Function Christopher D. Scharer,* Benjamin G. Barwick,* Benjamin A. Youngblood,*,† Rafi Ahmed,*,† and Jeremy M.
    [Show full text]
  • Negative Regulators of Colonic Peripheral Regulatory T Cell Development Teresa Lei Ai Washington University in St
    Washington University in St. Louis Washington University Open Scholarship Arts & Sciences Electronic Theses and Dissertations Arts & Sciences Spring 5-15-2018 Negative Regulators of Colonic Peripheral Regulatory T Cell Development Teresa Lei Ai Washington University in St. Louis Follow this and additional works at: https://openscholarship.wustl.edu/art_sci_etds Part of the Allergy and Immunology Commons, Immunology and Infectious Disease Commons, and the Medical Immunology Commons Recommended Citation Ai, Teresa Lei, "Negative Regulators of Colonic Peripheral Regulatory T Cell Development" (2018). Arts & Sciences Electronic Theses and Dissertations. 1509. https://openscholarship.wustl.edu/art_sci_etds/1509 This Dissertation is brought to you for free and open access by the Arts & Sciences at Washington University Open Scholarship. It has been accepted for inclusion in Arts & Sciences Electronic Theses and Dissertations by an authorized administrator of Washington University Open Scholarship. For more information, please contact [email protected]. WASHINGTON UNIVERSITY IN ST. LOUIS Division of Biology and Biomedical Sciences Immunology Dissertation Examination Committee: Chyi-Song Hsieh, Chair Paul M. Allen Kenneth M. Murphy Gwendalyn J. Randolph Wayne M. Yokoyama Negative Regulators of Colonic Peripheral Regulatory T Cell Development By Teresa Lei Ai A dissertation presented to The Graduate School of Washington University in partial fulfillment of the requirements for the degree of Doctor of Philosophy May 2018 St. Louis, Missouri
    [Show full text]