Quantitative Proteomic Analysis Reveals Caffeine-Perturbed Proteomic Profiles in Normal Bladder Epithelial Cells

Total Page:16

File Type:pdf, Size:1020Kb

Quantitative Proteomic Analysis Reveals Caffeine-Perturbed Proteomic Profiles in Normal Bladder Epithelial Cells HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Proteomics Manuscript Author . Author manuscript; Manuscript Author available in PMC 2019 October 11. Published in final edited form as: Proteomics. 2018 October ; 18(20): e1800190. doi:10.1002/pmic.201800190. Quantitative Proteomic Analysis Reveals Caffeine-Perturbed Proteomic Profiles in Normal Bladder Epithelial Cells Muhammad Shahid1, Minhyung Kim1, Austin Yeon1, Allen M. Andres2,3, Sungyong You1,4, and Jayoung Kim1,4,5,6,¶ 1Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA 2Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA, USA 3Cedars-Sinai Heart Institute, Los Angeles, California, CA, USA 4Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA 5University of California Los Angeles, CA, USA 6Department of Urology, Ga Cheon University College of Medicine, Incheon, South Korea Abstract Lower urinary tract symptoms (LUTS) are highly prevalent among the elderly and negatively impact quality-of-life. Since caffeinated beverages are enjoyed worldwide and the relationship between LUTS and caffeine is still not fully understood, it would be of particular interest to examine the underlying mechanisms that drive caffeine’s influence on LUTS development and progression. The aim of this study was to characterize the effects of caffeine on hTert immortalized normal bladder epithelial cells by investigating whether exposure to caffeine can cause potential changes in the bladder proteome and/or biological pathways. Labeled LC-MS/MS proteomic analysis found 57 proteins as being differentially expressed in caffeine-treated bladder epithelial cells, compared to controls; this included 32 upregulated and 25 downregulated proteins. Further functional gene enrichment analysis revealed that caffeine affected major biological pathways, including those for “muscle contraction” and “chromatin assembly”. These findings provide new scientific insights that may be useful in future studies investigating the role of caffeine in bladder dysfunctions. ¶Correspondence: Jayoung Kim, PhD., Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Davis 5071, 8700 Beverly Blvd., Los Angeles, CA 90048, Tel: +1-310-423-7168, Fax:+1-310-967-3809, [email protected]. Author contributions MS and AY involved and performed most of the experiments. SY and MK performed proteomics analysis and helped to draft paper. AA performed metabolic and biochemical analyses in vitro. JK designed and conceived the study and wrote the paper. All authors discussed the results and commented on the manuscript at all stages, and read and approved the manuscript. Conflicts of Interests The authors declare that there are no conflicts of interest regarding the publication of this paper. Shahid et al. Page 2 Author ManuscriptAuthor Keywords Manuscript Author Manuscript Author Manuscript Author Lower urinary tract symptoms; caffeine; global proteome; bladder epithelial cells; biological network INTRODUCTION It is estimated that by the end of 2018, 2.3 billion individuals will be affected by at least one lower urinary tract symptom (LUTS)[1]. These symptoms include urinary storage problems, such as urgency, frequency, nocturia, or voiding problems. LUTS also weighs heavily on overall quality of life; patients report significantly higher mental health issues, lower work productivity, and diminished general health status[2]. The current standard treatment for LUTS involves α-blockers, 5α-reductase inhibitors, and antimuscarinics[3]. However, these are mainly palliative and require consistent maintenance. Consequently, there is a substantial economic burden associated with LUTS[4]. Previous reports have demonstrated that diet and stress play important factors in the development and progression of LUTS[5]. In particular, caffeine, a naturally occurring compound, has been reported to be a potential dietary risk factor in developing LUTS[6]. Caffeine is ubiquitously found in many plants, including cocoa beans, tea leaves, and coffee beans. It is a stimulatory drug that is widely used to prevent sleepiness and can be found in over-the-counter medications, such as some pain remedies. It has also been observed that caffeine may aggravate or worsen urinary symptoms in patients who already have some form of LUTS[7]. In recent years, caffeinated drinks have become a staple of the average diet; more than 85% of adults in the U.S. regularly consume caffeine[8]. A longitudinal study of caffeine intake in young healthy volunteers found that subjects who regularly drank coffee had significant increases in urinary urgency and frequency[9]. Additionally, a separate study observed that greater coffee intake raised the odds of LUTS progression in men and women more than carbonated or citrus beverages[10]. The effects of caffeine have also been studied in the context of bladder cancer (BC); however, rather than finding a negative risk, there have been reported benefits. A case-control study of BC patients in Italy found no causal relationship between caffeine and BC[11]. A separate study found that caffeine may actually benefit BC patients by making cells more susceptible to chemotherapy and apoptosis[12]. Mechanistically, this has been reported to be mediated through caffeine’s effects on the tumor suppressor protein, p53[13]. Despite the potential link between caffeine and LUTS, research into causative mechanisms and functions is lacking. One prior study suggested that caffeine may be facilitating bladder instability and frequent urination by enhancing the activation of neuronal micturition centers through increased expression of transcription factor c-Fos and nerve growth factor[14]. While informative, this study focused primarily on the bladder muscles with little attention on the bladder epithelium, which is more anatomically exposed to urine and its biological/chemical contents. A separate study using a mouse model found that oral caffeine administration resulted in detrusor overactivity and increased bladder sensory signaling[15]. Further studies Proteomics. Author manuscript; available in PMC 2019 October 11. Shahid et al. Page 3 found similar effects on the detrusor muscle in humans[16]. However, the direct effects of Author ManuscriptAuthor Manuscript Author Manuscript Author Manuscript Author caffeine, its delivery into cells, and its mechanisms remain unestablished. Our present study sought to examine the cellular effects of caffeine on the bladder epithelium without any pathological conditions. Quantitative and global proteomics analysis found that caffeine can perturb the whole proteome, possibly through the regulation of chromatin assembly in normal bladder epithelial cells. MATERIALS AND METHODS Cell culture and cell proliferation assay. Immortalized normal human bladder epithelial cells, TRT-HU1, were maintained as described previously[17]. The TRT-HU1 cell line was constructed and extensively characterized in previously published papers. The passage number of each cell line was below 10, and mycoplasma contamination was tested for monthly via PCR analysis. Cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM), containing 10% fetal bovine serum (FBS, Invitrogen), 1% penicillin/streptomycin, and 1% L-glutamine (Sigma- Aldrich Corp., St. Louis, MO, USA) in a 37 °C humidified incubator with 5% CO2. To test cell growth during exposure to caffeine, TRT-HU1 cells were seeded onto 6-well plates with a density of 5 × 104 cells/well. Cells were then incubated with standard growth medium with varying doses of caffeine at 0, 0.005 mM, 0.05 mM, or 0.1 mM (C6035, Sigma-Aldrich Corp.) or vehicle for 24, 48, or 72 hrs. Cell proliferation was measured by manually counting cells using a hemocytometer. The averages of each count were used as the total density of the well after each time point. For crystal violet staining, the culture medium was removed, the cells were fixed with 4% paraformaldehyde at room temperature for 5 min and then stained with 0.05% crystal violet for 15 min. The cells were then washed with tap water, after which the water was removed and the cells were dried out on filter paper, and the cell plates were scanned and quantified as described previously[18]. All experiments were run in triplicate for each cell line, and the data are representative of three independent trials. Antibodies and reagents. Antibodies against various proteins were obtained from the following sources: ACTG2 (ab189385, Abcam), ACTA2 (ab5694, Abcam), MYH2 (ab124937, Abcam), MYH7B (ab172967, Abcam), HISTH2B (ab52599, Abcam), HIST1H2BM (SAB1301739, Sigma), and β-actin (A1978, Sigma). Commercially available horseradish peroxidase (HRP)- conjugated secondary antibodies (7074, 7076) were obtained from Cell Signaling Technology. All other chemical reagents were procured from Sigma Chemical Corp. Quantitative proteomics. Tandem mass tagging (TMT)-based quantitative proteomics was performed as described[19]. Briefly, cellular protein was extracted from caffeine-treated and control cells using 4% SDS- containing buffer. The protein concentration was measured using the Pierce 660nm Assay Kit. From each sample, 60 μg of protein was digested with trypsin using filter-aided sample preparation (FASP) and labeled with TMT6plex reagents in parallel. After TMT labeling, the peptides were merged, desalted with C18 spin columns (Thermo Scientific), and Proteomics. Author manuscript; available in PMC 2019 October
Recommended publications
  • Role of Phytochemicals in Colon Cancer Prevention: a Nutrigenomics Approach
    Role of phytochemicals in colon cancer prevention: a nutrigenomics approach Marjan J van Erk Promotor: Prof. Dr. P.J. van Bladeren Hoogleraar in de Toxicokinetiek en Biotransformatie Wageningen Universiteit Co-promotoren: Dr. Ir. J.M.M.J.G. Aarts Universitair Docent, Sectie Toxicologie Wageningen Universiteit Dr. Ir. B. van Ommen Senior Research Fellow Nutritional Systems Biology TNO Voeding, Zeist Promotiecommissie: Prof. Dr. P. Dolara University of Florence, Italy Prof. Dr. J.A.M. Leunissen Wageningen Universiteit Prof. Dr. J.C. Mathers University of Newcastle, United Kingdom Prof. Dr. M. Müller Wageningen Universiteit Dit onderzoek is uitgevoerd binnen de onderzoekschool VLAG Role of phytochemicals in colon cancer prevention: a nutrigenomics approach Marjan Jolanda van Erk Proefschrift ter verkrijging van graad van doctor op gezag van de rector magnificus van Wageningen Universiteit, Prof.Dr.Ir. L. Speelman, in het openbaar te verdedigen op vrijdag 1 oktober 2004 des namiddags te vier uur in de Aula Title Role of phytochemicals in colon cancer prevention: a nutrigenomics approach Author Marjan Jolanda van Erk Thesis Wageningen University, Wageningen, the Netherlands (2004) with abstract, with references, with summary in Dutch ISBN 90-8504-085-X ABSTRACT Role of phytochemicals in colon cancer prevention: a nutrigenomics approach Specific food compounds, especially from fruits and vegetables, may protect against development of colon cancer. In this thesis effects and mechanisms of various phytochemicals in relation to colon cancer prevention were studied through application of large-scale gene expression profiling. Expression measurement of thousands of genes can yield a more complete and in-depth insight into the mode of action of the compounds.
    [Show full text]
  • Histone-Related Genes Are Hypermethylated in Lung Cancer
    Published OnlineFirst October 1, 2019; DOI: 10.1158/0008-5472.CAN-19-1019 Cancer Genome and Epigenome Research Histone-Related Genes Are Hypermethylated in Lung Cancer and Hypermethylated HIST1H4F Could Serve as a Pan-Cancer Biomarker Shihua Dong1,Wei Li1, Lin Wang2, Jie Hu3,Yuanlin Song3, Baolong Zhang1, Xiaoguang Ren1, Shimeng Ji3, Jin Li1, Peng Xu1, Ying Liang1, Gang Chen4, Jia-Tao Lou2, and Wenqiang Yu1 Abstract Lung cancer is the leading cause of cancer-related deaths lated in all 17 tumor types from TCGA datasets (n ¼ 7,344), worldwide. Cytologic examination is the current "gold stan- which was further validated in nine different types of cancer dard" for lung cancer diagnosis, however, this has low sensi- (n ¼ 243). These results demonstrate that HIST1H4F can tivity. Here, we identified a typical methylation signature of function as a universal-cancer-only methylation (UCOM) histone genes in lung cancer by whole-genome DNA methyl- marker, which may aid in understanding general tumorigen- ation analysis, which was validated by The Cancer Genome esis and improve screening for early cancer diagnosis. Atlas (TCGA) lung cancer cohort (n ¼ 907) and was further confirmed in 265 bronchoalveolar lavage fluid samples with Significance: These findings identify a new biomarker for specificity and sensitivity of 96.7% and 87.0%, respectively. cancer detection and show that hypermethylation of histone- More importantly, HIST1H4F was universally hypermethy- related genes seems to persist across cancers. Introduction to its low specificity, LDCT is far from satisfactory as a screening tool for clinical application, similar to other currently used cancer Lung cancer is one of the most common malignant tumors and biomarkers, such as carcinoembryonic antigen (CEA), neuron- the leading cause of cancer-related deaths worldwide (1, 2).
    [Show full text]
  • Integrative Whole-Genome Sequence Analysis Reveals Roles of Regulatory
    www.nature.com/scientificreports OPEN Integrative whole-genome sequence analysis reveals roles of regulatory mutations in BCL6 and Received: 6 December 2016 Accepted: 27 June 2017 BCL2 in follicular lymphoma Published: xx xx xxxx Kirill Batmanov1, Wei Wang2, Magnar Bjørås2,4, Jan Delabie3 & Junbai Wang1 The contribution of mutations in regulatory regions to tumorigenesis has been the subject of many recent studies. We propose a new framework for integrative analysis of genome-wide sequencing data by considering diverse genetic information. This approach is applied to study follicular lymphoma (FL), a disease for which little is known about the contribution of regulatory gene mutations. Results from a test FL cohort revealed three novel highly recurrent regulatory mutation blocks near important genes implicated in FL, BCL6 and BCL2. Similar fndings were detected in a validation FL cohort. We also found transcription factors (TF) whose binding may be disturbed by these mutations in FL: disruption of FOX TF family near the BCL6 promoter may result in reduced BCL6 expression, which then increases BCL2 expression over that caused by BCL2 gene translocation. Knockdown experiments of two TF hits (FOXD2 or FOXD3) were performed in human B lymphocytes verifying that they modulate BCL6/BCL2 according to the computationally predicted efects of the SNVs on TF binding. Overall, our proposed integrative analysis facilitates non-coding driver identifcation and the new fndings may enhance the understanding of FL. High-throughput sequencing techniques have allowed researchers to study cancer-associated genetic and epi- genetic alterations in great detail. International consortia such as Te Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC) have sequenced thousands of samples of tumor and normal tissues, making it possible to survey the overall picture of molecular aberrations in cancer1, 2, as well as to inves- tigate particular mechanisms of oncogenesis3, 4.
    [Show full text]
  • Supplementary Materials Beryllium-Specific CD4+ T Cells
    Supplementary Materials Beryllium-specific CD4+ T cells induced by chemokine neoantigens perpetuate inflammation Michael T. Falta, Jeremy C. Crawford, Alex N. Tinega, Laurie G. Landry, Frances Crawford, Douglas G. Mack, Allison K. Martin, Shaikh M. Atif, Li Li, Radleigh G. Santos, Maki Nakayama, John W. Kappler, Lisa A. Maier, Paul G. Thomas, Clemencia Pinilla and Andrew P. Fontenot Supplementary Methods Figure S1. Cord diagrams of TR gene segment usage of individual CBD patient’s T cells. Figure S2. Beryllium and HLA-DP2 specificity of LKGGG CDR3β-expressing T cell hybridomas. Figure S3. IL-2 response of BAL-derived LKGGG CDR3β TCRs against an unbiased decapeptide PSL. Figure S4. IL-2 response of BAL-derived LKGGG CDR3β TCRs against a biased D5E8 decapeptide PSL. Figure S5. Identification of mimotopes that stimulate hybridomas expressing the LKGGG CDR3β motif. Figure S6. CCL4 peptide dose-response curves. Figure S7. Response of Be-specific non-LKGGG CDR3β motif TCRs to biometrical analysis naturally-occurring peptides. Figure S8. Investigation of chemokine/Be-specific TCRs potentially cross-reactive to plexin A/Be ligands. Table S1. TRA genes used by T cells expressing the LKGGG CDR3β motif. Table S2. Biometrical analysis peptides, their protein source and hybridoma IL-2 responses to peptide plus Be. Table S3. Mean EC50 values (nM) of two experiments for 4 hybridomas to CCL4 length variant peptides. Table S4. Mean EC50 values (nM) of two experiments for 4 hybridomas to CCL3 length variant peptides. Table S5. Demographics of CBD study population. 1 Supplementary Methods Generation of hybridomas expressing TCRs from human T cell clones.
    [Show full text]
  • Chromosome 21 Leading Edge Gene Set
    Chromosome 21 Leading Edge Gene Set Genes from chr21q22 that are part of the GSEA leading edge set identifying differences between trisomic and euploid samples. Multiple probe set IDs corresponding to a single gene symbol are combined as part of the GSEA analysis. Gene Symbol Probe Set IDs Gene Title 203865_s_at, 207999_s_at, 209979_at, adenosine deaminase, RNA-specific, B1 ADARB1 234539_at, 234799_at (RED1 homolog rat) UDP-Gal:betaGlcNAc beta 1,3- B3GALT5 206947_at galactosyltransferase, polypeptide 5 BACE2 217867_x_at, 222446_s_at beta-site APP-cleaving enzyme 2 1553227_s_at, 214820_at, 219280_at, 225446_at, 231860_at, 231960_at, bromodomain and WD repeat domain BRWD1 244622_at containing 1 C21orf121 240809_at chromosome 21 open reading frame 121 C21orf130 240068_at chromosome 21 open reading frame 130 C21orf22 1560881_a_at chromosome 21 open reading frame 22 C21orf29 1552570_at, 1555048_at_at, 1555049_at chromosome 21 open reading frame 29 C21orf33 202217_at, 210667_s_at chromosome 21 open reading frame 33 C21orf45 219004_s_at, 228597_at, 229671_s_at chromosome 21 open reading frame 45 C21orf51 1554430_at, 1554432_x_at, 228239_at chromosome 21 open reading frame 51 C21orf56 223360_at chromosome 21 open reading frame 56 C21orf59 218123_at, 244369_at chromosome 21 open reading frame 59 C21orf66 1555125_at, 218515_at, 221158_at chromosome 21 open reading frame 66 C21orf7 221211_s_at chromosome 21 open reading frame 7 C21orf77 220826_at chromosome 21 open reading frame 77 C21orf84 239968_at, 240589_at chromosome 21 open reading frame 84
    [Show full text]
  • Resveratrol Inhibits Cell Cycle Progression by Targeting Aurora Kinase a and Polo-Like Kinase 1 in Breast Cancer Cells
    3696 ONCOLOGY REPORTS 35: 3696-3704, 2016 Resveratrol inhibits cell cycle progression by targeting Aurora kinase A and Polo-like kinase 1 in breast cancer cells RUBICELI MEDINA-AGUILAR1, LAURENCE A. Marchat2, ELENA ARECHAGA OCAMPO3, Patricio GARIGLIO1, JAIME GARCÍA MENA1, NICOLÁS VILLEGAS SEPÚlveda4, MACARIO MartÍNEZ CASTILLO4 and CÉSAR LÓPEZ-CAMARILLO5 1Department of Genetics and Molecular Biology, CINVESTAV-IPN, Mexico D.F.; 2Molecular Biomedicine Program and Biotechnology Network, National School of Medicine and Homeopathy, National Polytechnic Institute, Mexico D.F.; 3Natural Sciences Department, Metropolitan Autonomous University, Mexico D.F.; 4Department of Molecular Biomedicine, CINVESTAV-IPN, Mexico D.F.; 5Oncogenomics and Cancer Proteomics Laboratory, Universidad Autónoma de la Ciudad de México, Mexico D.F., Mexico Received December 4, 2015; Accepted January 8, 2016 DOI: 10.3892/or.2016.4728 Abstract. The Aurora protein kinase (AURKA) and the MDA-MB-231 and MCF-7 cells. By western blot assays, we Polo-like kinase-1 (PLK1) activate the cell cycle, and they confirmed that resveratrol suppressed AURKA, CCND1 and are considered promising druggable targets in cancer CCNB1 at 24 and 48 h. In summary, we showed for the first time therapy. However, resistance to chemotherapy and to specific that resveratrol regulates cell cycle progression by targeting small-molecule inhibitors is common in cancer patients; thus AURKA and PLK1. Our findings highlight the potential use of alternative therapeutic approaches are needed to overcome resveratrol as an adjuvant therapy for breast cancer. clinical resistance. Here, we showed that the dietary compound resveratrol suppressed the cell cycle by targeting AURKA Introduction and PLK1 kinases. First, we identified genes modulated by resveratrol using a genome-wide analysis of gene expression Cancer development results from the interaction between in MDA-MB-231 breast cancer cells.
    [Show full text]
  • A High-Throughput Approach to Uncover Novel Roles of APOBEC2, a Functional Orphan of the AID/APOBEC Family
    Rockefeller University Digital Commons @ RU Student Theses and Dissertations 2018 A High-Throughput Approach to Uncover Novel Roles of APOBEC2, a Functional Orphan of the AID/APOBEC Family Linda Molla Follow this and additional works at: https://digitalcommons.rockefeller.edu/ student_theses_and_dissertations Part of the Life Sciences Commons A HIGH-THROUGHPUT APPROACH TO UNCOVER NOVEL ROLES OF APOBEC2, A FUNCTIONAL ORPHAN OF THE AID/APOBEC FAMILY A Thesis Presented to the Faculty of The Rockefeller University in Partial Fulfillment of the Requirements for the degree of Doctor of Philosophy by Linda Molla June 2018 © Copyright by Linda Molla 2018 A HIGH-THROUGHPUT APPROACH TO UNCOVER NOVEL ROLES OF APOBEC2, A FUNCTIONAL ORPHAN OF THE AID/APOBEC FAMILY Linda Molla, Ph.D. The Rockefeller University 2018 APOBEC2 is a member of the AID/APOBEC cytidine deaminase family of proteins. Unlike most of AID/APOBEC, however, APOBEC2’s function remains elusive. Previous research has implicated APOBEC2 in diverse organisms and cellular processes such as muscle biology (in Mus musculus), regeneration (in Danio rerio), and development (in Xenopus laevis). APOBEC2 has also been implicated in cancer. However the enzymatic activity, substrate or physiological target(s) of APOBEC2 are unknown. For this thesis, I have combined Next Generation Sequencing (NGS) techniques with state-of-the-art molecular biology to determine the physiological targets of APOBEC2. Using a cell culture muscle differentiation system, and RNA sequencing (RNA-Seq) by polyA capture, I demonstrated that unlike the AID/APOBEC family member APOBEC1, APOBEC2 is not an RNA editor. Using the same system combined with enhanced Reduced Representation Bisulfite Sequencing (eRRBS) analyses I showed that, unlike the AID/APOBEC family member AID, APOBEC2 does not act as a 5-methyl-C deaminase.
    [Show full text]
  • HIRA Orchestrates a Dynamic Chromatin Landscape in Senescence and Is Required for Suppression of Neoplasia
    Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press HIRA orchestrates a dynamic chromatin landscape in senescence and is required for suppression of neoplasia Taranjit Singh Rai,1,2,3 John J. Cole,1,2,5 David M. Nelson,1,2,5 Dina Dikovskaya,1,2 William J. Faller,1 Maria Grazia Vizioli,1,2 Rachael N. Hewitt,1,2 Orchi Anannya,1 Tony McBryan,1,2 Indrani Manoharan,1,2 John van Tuyn,1,2 Nicholas Morrice,1 Nikolay A. Pchelintsev,1,2 Andre Ivanov,1,2,4 Claire Brock,1,2 Mark E. Drotar,1,2 Colin Nixon,1 William Clark,1 Owen J. Sansom,1 Kurt I. Anderson,1 Ayala King,1 Karen Blyth,1 and Peter D. Adams1,2 1Beatson Institute for Cancer Research, Bearsden, Glasgow G61 1BD, United Kingdom; 2Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G61 1BD, United Kingdom; 3Institute of Biomedical and Environmental Health Research, University of West of Scotland, Paisley PA1 2BE, United Kingdom Cellular senescence is a stable proliferation arrest that suppresses tumorigenesis. Cellular senescence and associated tumor suppression depend on control of chromatin. Histone chaperone HIRA deposits variant histone H3.3 and histone H4 into chromatin in a DNA replication-independent manner. Appropriately for a DNA replication-independent chaperone, HIRA is involved in control of chromatin in nonproliferating senescent cells, although its role is poorly defined. Here, we show that nonproliferating senescent cells express and incorporate histone H3.3 and other canonical core histones into a dynamic chromatin landscape.
    [Show full text]
  • Down Syndrome Congenital Heart Disease: a Narrowed Region and a Candidate Gene Gillian M
    March/April 2001 ⅐ Vol. 3 ⅐ No. 2 article Down syndrome congenital heart disease: A narrowed region and a candidate gene Gillian M. Barlow, PhD1, Xiao-Ning Chen, MD1, Zheng Y. Shi, BS1, Gary E. Lyons, PhD2, David M. Kurnit, MD, PhD3, Livija Celle, MS4, Nancy B. Spinner, PhD4, Elaine Zackai, MD4, Mark J. Pettenati, PhD5, Alexander J. Van Riper, MS6, Michael J. Vekemans, MD7, Corey H. Mjaatvedt, PhD8, and Julie R. Korenberg, PhD, MD1 Purpose: Down syndrome (DS) is a major cause of congenital heart disease (CHD) and the most frequent known cause of atrioventricular septal defects (AVSDs). Molecular studies of rare individuals with CHD and partial duplications of chromosome 21 established a candidate region that included D21S55 through the telomere. We now report human molecular and cardiac data that narrow the DS-CHD region, excluding two candidate regions, and propose DSCAM (Down syndrome cell adhesion molecule) as a candidate gene. Methods: A panel of 19 individuals with partial trisomy 21 was evaluated using quantitative Southern blot dosage analysis and fluorescence in situ hybridization (FISH) with subsets of 32 BACs spanning the region defined by D21S16 (21q11.2) through the telomere. These BACs span the molecular markers D21S55, ERG, ETS2, MX1/2, collagen XVIII and collagen VI A1/A2. Fourteen individuals are duplicated for the candidate region, of whom eight (57%) have the characteristic spectrum of DS-CHD. Results: Combining the results from these eight individuals suggests the candidate region for DS-CHD is demarcated by D21S3 (defined by ventricular septal defect), through PFKL (defined by tetralogy of Fallot). Conclusions: These data suggest that the presence of three copies of gene(s) from the region is sufficient for the production of subsets of DS-CHD.
    [Show full text]
  • A Bacterial Protein Targets the BAHD1 Chromatin Complex to Stimulate Type III Interferon Response
    A bacterial protein targets the BAHD1 chromatin complex to stimulate type III interferon response Alice Lebreton, Goran Lakisic, Viviana Job, Lauriane Fritsch, To Nam Tham, Ana Camejo, Pierre-Jean Matteï, Béatrice Regnault, Marie-Anne Nahori, Didier Cabanes, et al. To cite this version: Alice Lebreton, Goran Lakisic, Viviana Job, Lauriane Fritsch, To Nam Tham, et al.. A bacterial protein targets the BAHD1 chromatin complex to stimulate type III interferon response. Science, American Association for the Advancement of Science, 2011, 331 (6022), pp.1319-21. 10.1126/sci- ence.1200120. cea-00819299 HAL Id: cea-00819299 https://hal-cea.archives-ouvertes.fr/cea-00819299 Submitted on 26 Jul 2020 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Lebreton et al. Science 2011 doi:10.1126/science.1200120 A Bacterial Protein Targets the BAHD1 Chromatin Complex to Stimulate Type III Interferon Response Alice Lebreton1,2,3, Goran Lakisic4, Viviana Job5, Lauriane Fritsch6, To Nam Tham1,2,3, Ana Camejo7, Pierre-Jean Matteï5, Béatrice Regnault8, Marie-Anne Nahori1,2,3, Didier Cabanes7, Alexis Gautreau4, Slimane Ait-Si-Ali6, Andréa Dessen5, Pascale Cossart1,2,3* and Hélène Bierne1,2,3* 1.
    [Show full text]
  • Histone Variants and Their Post-Translational Modifications in Primary Human Fat Cells
    Linköping University Post Print Histone Variants and Their Post-Translational Modifications in Primary Human Fat Cells Asa Jufvas, Peter Strålfors and Alexander Vener N.B.: When citing this work, cite the original article. Original Publication: Asa Jufvas, Peter Strålfors and Alexander Vener, Histone Variants and Their Post- Translational Modifications in Primary Human Fat Cells, 2011, PLOS ONE, (6), 1. http://dx.doi.org/10.1371/journal.pone.0015960 Licensee: Public Library of Science (PLoS) http://www.plos.org/ Postprint available at: Linköping University Electronic Press http://urn.kb.se/resolve?urn=urn:nbn:se:liu:diva-65933 Histone Variants and Their Post-Translational Modifications in Primary Human Fat Cells A˚ sa Jufvas, Peter Stra˚lfors, Alexander V. Vener* Department of Clinical and Experimental Medicine, Linko¨ping University, Linko¨ping, Sweden Abstract Epigenetic changes related to human disease cannot be fully addressed by studies of cells from cultures or from other mammals. We isolated human fat cells from subcutaneous abdominal fat tissue of female subjects and extracted histones from either purified nuclei or intact cells. Direct acid extraction of whole adipocytes was more efficient, yielding about 100 mg of protein with histone content of 60% –70% from 10 mL of fat cells. Differential proteolysis of the protein extracts by trypsin or ArgC-protease followed by nanoLC/MS/MS with alternating CID/ETD peptide sequencing identified 19 histone variants. Four variants were found at the protein level for the first time; particularly HIST2H4B was identified besides the only H4 isoform earlier known to be expressed in humans. Three of the found H2A potentially organize small nucleosomes in transcriptionally active chromatin, while two H2AFY variants inactivate X chromosome in female cells.
    [Show full text]
  • The Human Canonical Core Histone Catalogue David Miguel Susano Pinto*, Andrew Flaus*,†
    bioRxiv preprint doi: https://doi.org/10.1101/720235; this version posted July 30, 2019. 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 4.0 International license. The Human Canonical Core Histone Catalogue David Miguel Susano Pinto*, Andrew Flaus*,† Abstract Core histone proteins H2A, H2B, H3, and H4 are encoded by a large family of genes dis- tributed across the human genome. Canonical core histones contribute the majority of proteins to bulk chromatin packaging, and are encoded in 4 clusters by 65 coding genes comprising 17 for H2A, 18 for H2B, 15 for H3, and 15 for H4, along with at least 17 total pseudogenes. The canonical core histone genes display coding variation that gives rise to 11 H2A, 15 H2B, 4 H3, and 2 H4 unique protein isoforms. Although histone proteins are highly conserved overall, these isoforms represent a surprising and seldom recognised variation with amino acid identity as low as 77 % between canonical histone proteins of the same type. The gene sequence and protein isoform diversity also exceeds com- monly used subtype designations such as H2A.1 and H3.1, and exists in parallel with the well-known specialisation of variant histone proteins. RNA sequencing of histone transcripts shows evidence for differential expression of histone genes but the functional significance of this variation has not yet been investigated. To assist understanding of the implications of histone gene and protein diversity we have catalogued the entire human canonical core histone gene and protein complement.
    [Show full text]