Functional Characterization of Thousands of Type 2 Diabetes-Associated And

Total Page:16

File Type:pdf, Size:1020Kb

Functional Characterization of Thousands of Type 2 Diabetes-Associated And bioRxiv preprint doi: https://doi.org/10.1101/2020.02.12.939348; this version posted February 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Functional characterization of thousands of type 2 diabetes-associated and 2 chromatin-modulating variants under steady state and endoplasmic reticulum 3 stress 4 5 Shubham Khetan1,3, Susan Kales2, Romy Kursawe1, Alexandria Jillette1, Steven K. 6 Reilly4, Duygu Ucar1,3,5, Ryan Tewhey2,6,7*, and Michael L. Stitzel1,3,5* 7 8 1The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA 9 2The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, 04609, USA 10 3Department of Genetics and Genome Sciences, University of Connecticut, Farmington, 11 CT, 06032, USA, 12 4Broad Institute of MIT and Harvard, Cambridge, MA, USA 13 5Institute of Systems Genomics, University of Connecticut, Farmington, CT, 06032, USA 14 6Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, 15 ME, 04469, USA. 16 7Tufts University School of Medicine, Boston, MA, 02111, USA 17 18 *Corresponding authors: [email protected] and [email protected] 19 20 Keywords: Massively Parallel Reporter Assay (MPRA), human pancreatic islets, MIN6 21 beta cells, chromatin accessibility, ATAC-seq, chromatin accessibility quantitative trait 22 locus (caQTLs), genome-wide association study (GWAS), single nucleotide 23 polymorphism (SNP), Type 2 diabetes (T2D), endoplasmic reticulum (ER) stress, short 24 interspersed nuclear elements (SINE), Alu elements. bioRxiv preprint doi: https://doi.org/10.1101/2020.02.12.939348; this version posted February 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 25 Abstract: (199 words) 26 A major goal in functional genomics and complex disease genetics is to identify 27 functional cis-regulatory elements (CREs) and single nucleotide polymorphisms (SNPs) 28 altering CRE activity in disease-relevant cell types and environmental conditions. We 29 tested >13,000 sequences containing each allele of 6,628 SNPs associated with altered 30 in vivo chromatin accessibility in human islets and/or type 2 diabetes risk (T2D GWAS 31 SNPs) for transcriptional activity in ß cell under steady state and endoplasmic reticulum 32 (ER) stress conditions using the massively parallel reporter assay (MPRA). 33 Approximately 30% (n=1,983) of putative CREs were active in at least one condition. 34 SNP allelic effects on in vitro MPRA activity strongly correlated with their effects on in 35 vivo islet chromatin accessibility (Pearson r=0.52), i.e., alleles associated with increased 36 chromatin accessibility exhibited higher MPRA activity. Importantly, MPRA identified 37 220/2500 T2D GWAS SNPs, representing 104 distinct association signals, that 38 significantly altered transcriptional activity in ß cells. This study has thus identified 39 functional ß cell transcription-activating sequences with in vivo relevance, uncovered 40 regulatory features that modulate transcriptional activity in ß cells under steady state and 41 ER stress conditions, and substantially expanded the set of putative functional variants 42 that modulate transcriptional activity in ß cells from thousands of genetically-linked T2D 43 GWAS SNPs. bioRxiv preprint doi: https://doi.org/10.1101/2020.02.12.939348; this version posted February 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 44 Introduction 45 Studies over the past decade have identified millions of single nucleotide 46 polymorphisms (SNPs) in the human population1. Genome-wide association studies 47 (GWAS) have linked thousands of these SNPs to variability in physiological traits and 48 disease susceptibility2, including type 2 diabetes (T2D)3. The overwhelming majority of 49 GWAS SNPs reside in non-coding, regulatory regions of the genome3,4, implicating 50 altered transcriptional regulation as a common molecular mechanism underlying disease 51 risk. Compared to protein-coding regions, predicting regulatory functions of non-coding 52 sequences, and the effect of SNPs within them, remains a significant challenge. 53 Approaches such as ATAC-seq identify regions of accessible chromatin as a 54 marker of in vivo cis-regulatory elements (CREs)5. However, CREs identified by 55 chromatin accessibility (ATAC-seq peaks) vary significantly in length. Moreover, ATAC- 56 seq peaks are characterized by 1 or more summits, which have higher chromatin 57 accessibility than flanking genomic regions also within ATAC-seq peaks6. Few studies 58 have explored the relationship between transcriptional activity and open chromatin 59 regions defined by ATAC-seq7,8. For instance, it is not known whether higher chromatin 60 accessibility within ATAC-seq peaks also corresponds to enhanced transcriptional 61 activity, or whether proximity to ATAC-seq peak summits influences the likelihood for a 62 SNP to affect enhancer activity. Studies designed to identify chromatin accessibility 63 quantitative trait loci (caQTL) are being increasingly applied to nominate SNPs altering 64 CRE activity9,10. Previously, we identified 2,949 caQTLs in human islets11, which were 65 significantly enriched for type 2 diabetes (T2D)-associated SNPs, highlighting the utility 66 of this approach to nominate putative disease-associated SNPs affecting CRE activity in 67 relevant cell types. However, caQTLs are correlative associations, and high throughput 68 assays to directly test variant effects are needed to experimentally establish causality. 69 bioRxiv preprint doi: https://doi.org/10.1101/2020.02.12.939348; this version posted February 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 70 Massively parallel reporter assays (MPRAs) have been developed as a functional 71 genomics platform to interrogate the enhancer potential of thousands of sequences 72 simultaneously12 under a variety of (patho)physiologic conditions. By introducing 73 nucleotide changes in a given sequence of interest, the effect of naturally occurring 74 sequence variants in the human population on MPRA activity can also be elucidated13. 75 Several studies have employed MPRA to identify functional SNPs associated with red 76 blood cell traits14, adiposity15, osteoarthritis16, cancer and eQTLs13. 77 Although studies over the past several years implicate altered islet CRE function 78 in T2D genetic risk and progression, they have colocalized only ~1/4 of T2D-associated 79 loci to altered chromatin accessibility and/or gene expression levels in islets11,17–20. We 80 hypothesize that this is partially because previous studies measured chromatin 81 accessibility (caQTLs) and gene expression (eQTL) in islets cultured under steady state 82 conditions11,21, consequently missing important gene-environment interactions 83 characteristic of a complex disorder like T2D. For example, endoplasmic reticulum (ER) 84 stress, which is elicited by the high demands of insulin production and secretion on 85 protein folding capacity in β cells, leads to (patho)physiologic adaptations. Low levels of 86 ER stress are a signal for β cells to proliferate as a mechanism to meet higher insulin 87 secretion demands22. However, uncompensated ER stress induced by genetic and/or 88 environmental perturbations leads to β cell failure and T2D23. 89 Here, we applied MPRA to directly test >13,000 sequences, including those 90 overlapping T2D GWAS and islet caQTL SNPs, for their ability to activate ß cell 91 transcription in steady state conditions and after exposure to the ER stress-inducing 92 agent thapsigargin or DMSO solvent control. We identified motifs and features of 93 sequences affecting MPRA activity in each condition and linked allelic effects on MPRA 94 activity to T2D genetics and in vivo islet chromatin accessibility. These results 95 demonstrate the power of MPRA to elucidate in vivo relevant ß cell transcriptional bioRxiv preprint doi: https://doi.org/10.1101/2020.02.12.939348; this version posted February 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 96 activating sequences and define the regulatory grammar of these sequences under 97 steady state and stress-responsive conditions. We nominate 54 putative causal variants 98 among multiple genetically-linked islet dysfunction and T2D GWAS SNPs for further 99 study. 100 101 Results 102 Selection and testing of sequences for MPRA activity in β cells 103 To test putative islet β cell regulatory sequences for their ability to enhance 104 transcription from a minimal promoter and to identify SNPs that alter regulatory activity, 105 we generated an MPRA library containing: i) 1,910 SNPs significantly associated with 106 changes in human islet chromatin accessibility (caQTLs)11; ii) 2,218 control SNPs 107 overlapping human islet ATAC-seq peaks, which were reported to have no correlation 108 with changes in human islet chromatin accessibility11; and iii) 2,500 index and genetically 109 linked (r2>0.8) SNPs/indels corresponding to 259 T2D association signals in the 110 NHGRI/EBI GWAS Catalogue (Figure 1a,
Recommended publications
  • Mouse Fscn3 Conditional Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Fscn3 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Fscn3 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Fscn3 gene (NCBI Reference Sequence: NM_019569 ; Ensembl: ENSMUSG00000029707 ) is located on Mouse chromosome 6. 7 exons are identified, with the ATG start codon in exon 1 and the TAG stop codon in exon 6 (Transcript: ENSMUST00000031719). Exon 2 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Fscn3 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP24-176I9 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 2 starts from about 9.71% of the coding region. The knockout of Exon 2 will result in frameshift of the gene. The size of intron 1 for 5'-loxP site insertion: 1817 bp, and the size of intron 2 for 3'-loxP site insertion: 839 bp. The size of effective cKO region: ~1197 bp. The cKO region does not have any other known gene. Page 1 of 7 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 2 3 7 Targeting vector Targeted allele Constitutive KO allele (After Cre recombination) Legends Homology arm Exon of mouse Fscn3 cKO region loxP site Page 2 of 7 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • Protein Interaction Network of Alternatively Spliced Isoforms from Brain Links Genetic Risk Factors for Autism
    ARTICLE Received 24 Aug 2013 | Accepted 14 Mar 2014 | Published 11 Apr 2014 DOI: 10.1038/ncomms4650 OPEN Protein interaction network of alternatively spliced isoforms from brain links genetic risk factors for autism Roser Corominas1,*, Xinping Yang2,3,*, Guan Ning Lin1,*, Shuli Kang1,*, Yun Shen2,3, Lila Ghamsari2,3,w, Martin Broly2,3, Maria Rodriguez2,3, Stanley Tam2,3, Shelly A. Trigg2,3,w, Changyu Fan2,3, Song Yi2,3, Murat Tasan4, Irma Lemmens5, Xingyan Kuang6, Nan Zhao6, Dheeraj Malhotra7, Jacob J. Michaelson7,w, Vladimir Vacic8, Michael A. Calderwood2,3, Frederick P. Roth2,3,4, Jan Tavernier5, Steve Horvath9, Kourosh Salehi-Ashtiani2,3,w, Dmitry Korkin6, Jonathan Sebat7, David E. Hill2,3, Tong Hao2,3, Marc Vidal2,3 & Lilia M. Iakoucheva1 Increased risk for autism spectrum disorders (ASD) is attributed to hundreds of genetic loci. The convergence of ASD variants have been investigated using various approaches, including protein interactions extracted from the published literature. However, these datasets are frequently incomplete, carry biases and are limited to interactions of a single splicing isoform, which may not be expressed in the disease-relevant tissue. Here we introduce a new interactome mapping approach by experimentally identifying interactions between brain-expressed alternatively spliced variants of ASD risk factors. The Autism Spliceform Interaction Network reveals that almost half of the detected interactions and about 30% of the newly identified interacting partners represent contribution from splicing variants, emphasizing the importance of isoform networks. Isoform interactions greatly contribute to establishing direct physical connections between proteins from the de novo autism CNVs. Our findings demonstrate the critical role of spliceform networks for translating genetic knowledge into a better understanding of human diseases.
    [Show full text]
  • Associated Marks Are Enriched in Imprinted Gene Regions and Predict Allele-Specific Modification
    Downloaded from genome.cshlp.org on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press Methods Overlapping euchromatin/heterochromatin- associated marks are enriched in imprinted gene regions and predict allele-specific modification Bo Wen,1,4 Hao Wu,2,4 Hans Bjornsson,1 Roland D. Green,3 Rafael Irizarry,2 and Andrew P. Feinberg1,5 1Department of Medicine and Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; 2Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA; 3NimbleGen Systems, Inc., Madison, Wisconsin 53711, USA Most genome-level analysis treats the two parental alleles equivalently, yet diploid genomes contain two parental genomes that are often epigenetically distinct. While single nucleotide polymorphisms (SNPs) can be used to distinguish these genomes, it would be useful to develop a generalized strategy for identifying candidate genes or regions showing allele-specific differences, independent of SNPs. We have explored this problem by looking for overlapping marks in the genome related to both euchromatin (histone H3 dimethyl lysine-4 [H3K4Me2]) and heterochromatin (DNA methylation [DNAm]). “Double hits” were defined by the intersection of H3K4Me2 and DNAm. For the top 5% of marks, defined by a sliding window, imprinted gene regions were enriched for double hits 5.4-fold. When the location information of CTCF binding sites were integrated, the “triple hits” were enriched 76-fold for known imprinted genes in the regions studied. The double hits in imprinted genes were found to occur usually at the site of alternative or antisense transcripts. In addition, four of four imprinted genes tested showing double hits also showed allele-specific methylation.
    [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]
  • Noelia Díaz Blanco
    Effects of environmental factors on the gonadal transcriptome of European sea bass (Dicentrarchus labrax), juvenile growth and sex ratios Noelia Díaz Blanco Ph.D. thesis 2014 Submitted in partial fulfillment of the requirements for the Ph.D. degree from the Universitat Pompeu Fabra (UPF). This work has been carried out at the Group of Biology of Reproduction (GBR), at the Department of Renewable Marine Resources of the Institute of Marine Sciences (ICM-CSIC). Thesis supervisor: Dr. Francesc Piferrer Professor d’Investigació Institut de Ciències del Mar (ICM-CSIC) i ii A mis padres A Xavi iii iv Acknowledgements This thesis has been made possible by the support of many people who in one way or another, many times unknowingly, gave me the strength to overcome this "long and winding road". First of all, I would like to thank my supervisor, Dr. Francesc Piferrer, for his patience, guidance and wise advice throughout all this Ph.D. experience. But above all, for the trust he placed on me almost seven years ago when he offered me the opportunity to be part of his team. Thanks also for teaching me how to question always everything, for sharing with me your enthusiasm for science and for giving me the opportunity of learning from you by participating in many projects, collaborations and scientific meetings. I am also thankful to my colleagues (former and present Group of Biology of Reproduction members) for your support and encouragement throughout this journey. To the “exGBRs”, thanks for helping me with my first steps into this world. Working as an undergrad with you Dr.
    [Show full text]
  • Supplementary Data
    Supplementary Fig. 1 A B Responder_Xenograft_ Responder_Xenograft_ NON- NON- Lu7336, Vehicle vs Lu7466, Vehicle vs Responder_Xenograft_ Responder_Xenograft_ Sagopilone, Welch- Sagopilone, Welch- Lu7187, Vehicle vs Lu7406, Vehicle vs Test: 638 Test: 600 Sagopilone, Welch- Sagopilone, Welch- Test: 468 Test: 482 Responder_Xenograft_ NON- Lu7860, Vehicle vs Responder_Xenograft_ Sagopilone, Welch - Lu7558, Vehicle vs Test: 605 Sagopilone, Welch- Test: 333 Supplementary Fig. 2 Supplementary Fig. 3 Supplementary Figure S1. Venn diagrams comparing probe sets regulated by Sagopilone treatment (10mg/kg for 24h) between individual models (Welsh Test ellipse p-value<0.001 or 5-fold change). A Sagopilone responder models, B Sagopilone non-responder models. Supplementary Figure S2. Pathway analysis of genes regulated by Sagopilone treatment in responder xenograft models 24h after Sagopilone treatment by GeneGo Metacore; the most significant pathway map representing cell cycle/spindle assembly and chromosome separation is shown, genes upregulated by Sagopilone treatment are marked with red thermometers. Supplementary Figure S3. GeneGo Metacore pathway analysis of genes differentially expressed between Sagopilone Responder and Non-Responder models displaying –log(p-Values) of most significant pathway maps. Supplementary Tables Supplementary Table 1. Response and activity in 22 non-small-cell lung cancer (NSCLC) xenograft models after treatment with Sagopilone and other cytotoxic agents commonly used in the management of NSCLC Tumor Model Response type
    [Show full text]
  • PWWP2A Binds Distinct Chromatin Moieties and Interacts with an MTA1-Specific Core Nurd Complex
    ARTICLE DOI: 10.1038/s41467-018-06665-5 OPEN PWWP2A binds distinct chromatin moieties and interacts with an MTA1-specific core NuRD complex Stephanie Link1,2, Ramona M.M. Spitzer1,2, Maryam Sana3, Mario Torrado3, Moritz C. Völker-Albert1, Eva C. Keilhauer4,9, Thomas Burgold5,10, Sebastian Pünzeler1,11, Jason K.K. Low3, Ida Lindström 3, Andrea Nist6, Catherine Regnard1, Thorsten Stiewe 6,7, Brian Hendrich5, Axel Imhof 1,8, Matthias Mann 4,8, Joel P. Mackay 3, Marek Bartkuhn2 & Sandra B. Hake 2,8 1234567890():,; Chromatin structure and function is regulated by reader proteins recognizing histone mod- ifications and/or histone variants. We recently identified that PWWP2A tightly binds to H2A. Z-containing nucleosomes and is involved in mitotic progression and cranial–facial devel- opment. Here, using in vitro assays, we show that distinct domains of PWWP2A mediate binding to free linker DNA as well as H3K36me3 nucleosomes. In vivo, PWWP2A strongly recognizes H2A.Z-containing regulatory regions and weakly binds H3K36me3-containing gene bodies. Further, PWWP2A binds to an MTA1-specific subcomplex of the NuRD complex (M1HR), which consists solely of MTA1, HDAC1, and RBBP4/7, and excludes CHD, GATAD2 and MBD proteins. Depletion of PWWP2A leads to an increase of acetylation levels on H3K27 as well as H2A.Z, presumably by impaired chromatin recruitment of M1HR. Thus, this study identifies PWWP2A as a complex chromatin-binding protein that serves to direct the deacetylase complex M1HR to H2A.Z-containing chromatin, thereby promoting changes in histone acetylation levels. 1 Department of Molecular Biology, BioMedical Center (BMC), Ludwig-Maximilians-University Munich, 82152 Planegg-Martinsried, Germany.
    [Show full text]
  • Chapter 14: Genetics of Type 2 Diabetes
    CHAPTER 14 GENETICS OF TYPE 2 DIABETES Jose C. Florez, MD, PhD, Miriam S. Udler, MD, PhD, and Robert L. Hanson, MD, MPH Dr. Jose C. Florez is Chief of the Diabetes Unit and an investigator in the Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, and Co-Director of the Program in Metabolism and Institute Member in the Broad Institute, Cambridge, MA, and Associate Professor in the Department of Medicine, Harvard Medical School, Boston, MA. Dr. Miriam S. Udler is Clinical Fellow in the Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, and Postdoctoral Fellow in the Programs in Metabolism and Medical & Population Genetics, Broad Institute, Cambridge, MA, and Research Fellow in the Department of Medicine, Harvard Medical School, Boston, MA. Dr. Robert L. Hanson is Clinical Investigator and Head, Genetic Epidemiology and Statistics Unit in the Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ. SUMMARY Type 2 diabetes is thought to result from a has been found to have a stronger effect insufficient sample sizes to detect small combination of environmental, behavioral, than the rs7903146 SNP in TCF7L2, which effects, a nearly exclusive focus on popu- and genetic factors, with the heritability itself has only a modest effect (odds ratio lations of European descent, an imperfect of type 2 diabetes estimated to be in the ~1.4). Nonetheless, GWAS findings have capture of uncommon genetic variants, range of 25% to 72% based on family and illustrated novel pathways, pointed toward an incomplete ascertainment of alternate twin studies.
    [Show full text]
  • WO 2016/040794 Al 17 March 2016 (17.03.2016) P O P C T
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2016/040794 Al 17 March 2016 (17.03.2016) P O P C T (51) International Patent Classification: AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, C12N 1/19 (2006.01) C12Q 1/02 (2006.01) BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, C12N 15/81 (2006.01) C07K 14/47 (2006.01) DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IR, IS, JP, KE, KG, KN, KP, KR, (21) International Application Number: KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, MG, PCT/US20 15/049674 MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, (22) International Filing Date: PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, SC, 11 September 2015 ( 11.09.201 5) SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. (25) Filing Language: English (84) Designated States (unless otherwise indicated, for every (26) Publication Language: English kind of regional protection available): ARIPO (BW, GH, (30) Priority Data: GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, 62/050,045 12 September 2014 (12.09.2014) US TZ, UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ, TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, (71) Applicant: WHITEHEAD INSTITUTE FOR BIOMED¬ DK, EE, ES, FI, FR, GB, GR, HR, HU, IE, IS, IT, LT, LU, ICAL RESEARCH [US/US]; Nine Cambridge Center, LV, MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, Cambridge, Massachusetts 02142-1479 (US).
    [Show full text]
  • Global Patterns of Changes in the Gene Expression Associated with Genesis of Cancer a Dissertation Submitted in Partial Fulfillm
    Global Patterns Of Changes In The Gene Expression Associated With Genesis Of Cancer A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy at George Mason University By Ganiraju Manyam Master of Science IIIT-Hyderabad, 2004 Bachelor of Engineering Bharatiar University, 2002 Director: Dr. Ancha Baranova, Associate Professor Department of Molecular & Microbiology Fall Semester 2009 George Mason University Fairfax, VA Copyright: 2009 Ganiraju Manyam All Rights Reserved ii DEDICATION To my parents Pattabhi Ramanna and Veera Venkata Satyavathi who introduced me to the joy of learning. To friends, family and colleagues who have contributed in work, thought, and support to this project. iii ACKNOWLEDGEMENTS I would like to thank my advisor, Dr. Ancha Baranova, whose tolerance, patience, guidance and encouragement helped me throughout the study. This dissertation would not have been possible without her ever ending support. She is very sincere and generous with her knowledge, availability, compassion, wisdom and feedback. I would also like to thank Dr. Vikas Chandhoke for funding my research generously during my doctoral study at George Mason University. Special thanks go to Dr. Patrick Gillevet, Dr. Alessandro Giuliani, Dr. Maria Stepanova who devoted their time to provide me with their valuable contributions and guidance to formulate this project. Thanks to the faculty of Molecular and Micro Biology (MMB) department, Dr. Jim Willett and Dr. Monique Vanhoek in embedding valuable thoughts to this dissertation by being in my dissertation committee. I would also like to thank the present and previous doctoral program directors, Dr. Daniel Cox and Dr. Geraldine Grant, for facilitating, allowing, and encouraging me to work in this project.
    [Show full text]
  • Recent Progress in Genetic and Epigenetic Research on Type 2 Diabetes
    OPEN Experimental & Molecular Medicine (2016) 48, e220; doi:10.1038/emm.2016.7 & 2016 KSBMB. All rights reserved 2092-6413/16 www.nature.com/emm REVIEW Recent progress in genetic and epigenetic research on type 2 diabetes Soo Heon Kwak1 and Kyong Soo Park1,2,3 Type 2 diabetes (T2DM) is a common complex metabolic disorder that has a strong genetic predisposition. During the past decade, progress in genetic association studies has enabled the identification of at least 75 independent genetic loci for T2DM, thus allowing a better understanding of the genetic architecture of T2DM. International collaborations and large-scale meta- analyses of genome-wide association studies have made these achievements possible. However, whether the identified common variants are causal is largely unknown. In addition, the detailed mechanism of how these genetic variants exert their effect on the pathogenesis of T2DM requires further investigation. Currently, there are ongoing large-scale sequencing studies to identify rare, functional variants for T2DM. Environmental factors also have a crucial role in the development of T2DM. These could modulate gene expression via epigenetic mechanisms, including DNA methylation, histone modification and microRNA regulation. There is evidence that epigenetic changes are important in the development of T2DM. Recent studies have identified several DNA methylation markers of T2DM from peripheral blood and pancreatic islets. In this review, we will briefly summarize the recent progress in the genetic and epigenetic research on
    [Show full text]
  • Pure Duplication of the Distal Long Arm of Chromosome 15 with Ebstein Anomaly and Clavicular Anomaly
    Hindawi Publishing Corporation Case Reports in Genetics Volume 2011, Article ID 898706, 5 pages doi:10.1155/2011/898706 Case Report Pure Duplication of the Distal Long Arm of Chromosome 15 with Ebstein Anomaly and Clavicular Anomaly Rachel O’Connor,1 Amel Al-Murrani,1 Salim Aftimos,2 Philip Asquith,1 Roberto Mazzaschi,1 Dominique Eyrolle-Guignot,3 Alice M. George,1 and Donald R. Love1, 4 1 Diagnostic Genetics, LabPlus, Auckland City Hospital, P.O. Box 110031, Auckland 1148, New Zealand 2 Northern Regional Genetic Service, Auckland City Hospital, Private Bag 92024, Auckland 1142, New Zealand 3 Maternit´e Andrea De Balmain, Centre Hospitalier du Taaone, BP 1640, 98 713 Papeete, Polyn´esie Franc¸aise, France 4 School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand Correspondence should be addressed to Donald R. Love, [email protected] Received 24 June 2011; Accepted 21 July 2011 Academic Editors: D. J. Bunyan, P. D. Cotter, and G. Velagaleti Copyright © 2011 Rachel O’Connor et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. This report is of a patient with pure trisomy of 15q24-qter who presents with the rare Ebstein anomaly and a previously unreported skeletal anomaly. Chromosome microarray analysis allowed high-resolution identification of the extent of the trisomy and provided a means of achieving higher-resolution breakpoint data. The phenotypic expression of unbalanced chromosomal regions is a complex phenomenon, and fine mapping of the involved region, as described here, is only a first step on the path to its full understanding.
    [Show full text]