Virtual Chip-Seq: Predicting Transcription Factor Binding

Total Page:16

File Type:pdf, Size:1020Kb

Virtual Chip-Seq: Predicting Transcription Factor Binding bioRxiv preprint doi: https://doi.org/10.1101/168419; this version posted March 12, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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 Virtual ChIP-seq: predicting transcription factor binding 2 by learning from the transcriptome 1,2,3 1,2,3,4,5 3 Mehran Karimzadeh and Michael M. Hoffman 1 4 Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada 2 5 Princess Margaret Cancer Centre, Toronto, ON, Canada 3 6 Vector Institute, Toronto, ON, Canada 4 7 Department of Computer Science, University of Toronto, Toronto, ON, Canada 5 8 Lead contact: michael.hoff[email protected] 9 March 8, 2019 10 Abstract 11 Motivation: 12 Identifying transcription factor binding sites is the first step in pinpointing non-coding mutations 13 that disrupt the regulatory function of transcription factors and promote disease. ChIP-seq is 14 the most common method for identifying binding sites, but performing it on patient samples is 15 hampered by the amount of available biological material and the cost of the experiment. Existing 16 methods for computational prediction of regulatory elements primarily predict binding in genomic 17 regions with sequence similarity to known transcription factor sequence preferences. This has limited 18 efficacy since most binding sites do not resemble known transcription factor sequence motifs, and 19 many transcription factors are not even sequence-specific. 20 Results: 21 We developed Virtual ChIP-seq, which predicts binding of individual transcription factors in new 22 cell types using an artificial neural network that integrates ChIP-seq results from other cell types 23 and chromatin accessibility data in the new cell type. Virtual ChIP-seq also uses learned associ- 24 ations between gene expression and transcription factor binding at specific genomic regions. This 25 approach outperforms methods that predict TF binding solely based on sequence preference, pre- 26 dicting binding for 36 transcription factors (Matthews correlation coefficient > 0:3). 27 Availability: 28 The datasets we used for training and validation are available at https://virchip.hoffmanlab. 29 org. We have deposited in Zenodo the current version of our software (http://doi.org/10. 30 5281/zenodo.1066928), datasets (http://doi.org/10.5281/zenodo.823297), predictions for 36 31 transcription factors on Roadmap Epigenomics cell types (http://doi.org/10.5281/zenodo. 32 1455759), and predictions in Cistrome as well as ENCODE-DREAM in vivo TF Binding Site 33 Prediction Challenge (http://doi.org/10.5281/zenodo.1209308). 1 bioRxiv preprint doi: https://doi.org/10.1101/168419; this version posted March 12, 2019. 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. 34 1 Introduction 35 Transcription factor (TF) binding regulates gene expression. Each TF can harmonize expression of 36 many genes by binding to genomic regions that regulate transcription. Cellular machinery utilizes 37 these master regulators to regulate key cellular processes and adapt to environmental stimuli. 38 Alteration in sequence or quantity of a given TF can impact expression of many genes. In fact, 39 these alterations can be the primary cause of hereditary disorders, complex disease, autoimmune 1 40 defects, and cancer . 41 TFs bind to accessible chromatin based on weak non-covalent interactions between amino acid 2 3 42 residues and nucleic acids. DNA's primary structure (sequence) , secondary structure (shape) , and 4 43 tertiary structure (conformation) all play roles in TF binding. Many TFs form a complex with 44 others as well as chromatin-binding proteins and therefore bind to DNA indirectly. Some TFs also 45 have different isoforms and undergo various post-translational modifications. In vitro assays, such 5 46 as high throughput systematic evolution of ligands by exponential enrichment (HT-SELEX) and 6 47 protein binding microarrays , have provided a compelling understanding of context-independent 7 48 TF sequence and shape preference . Yet, for the aforementioned reasons, performance of models 8,9 49 trained on these in vitro data are poor when applied on in vivo experiments . To address this 50 challenge, we must explore how to better model DNA shape, TF-TF interactions, and context- 51 dependent TF binding. 10 52 Chromatin immunoprecipitation and sequencing (ChIP-seq) and similar methods, such as 11 12 53 ChIP-exo and ChIP-nexus , can map the presence of a given TF in the genome of a biological 54 sample. To map TFs, these assays require a minimum of 1,000,000 to 100,000,000 cells, depending 55 on properties of the TF itself and available antibodies. Such large numbers of cells are not often 56 available from clinical samples. Therefore, it is impossible to systematically assess TF binding in 57 most disease systems. Assessing chromatin accessibility through transposase-accessible chromatin 13 58 using sequencing (ATAC-seq) , however, requires only hundreds or thousands of cells. One can 59 obtain this many cells from many more clinical samples. While chromatin accessibility does not de- 60 termine TF binding, several methods use this information together with knowledge of TF sequence 14,15,16 61 preference, genomic conservation, and other genomic features to predict TF binding . 62 Predicting TF binding with motif discovery tools within chromatin accessible regions has helped 17 63 us understand the role of several TFs in various disease. For example, He et al. used motif discov- 64 ery tools to identify the role of OCT1 and NKX3-1 after prolonged androgen stimulation in prostate 18 65 cancer. Similarly, Bailey et al. discovered that a known breast cancer risk single nucleotide poly- 66 morphism (SNP) upstream of ESR1 disrupts GATA3 binding and enhances expression of ESR1. 67 We propose that using more accurate tools to predict TF binding will allow understanding the role 68 of TF binding in more contexts. 69 Previous studies have used various approaches to predict TF binding. Several methods use unsu- 14 15 70 pervised approaches such as hierarchical mixture models or hidden Markov models to identify 71 transcription factor footprint using chromatin accessibility data. These approaches use sequence 72 motif scores to attribute footprints to different transcription factors. Convolutional neural network 73 models can boost precision by learning sequence preferences from in vivo, rather than in vitro 20,21 74 data . Variation in sequence specificity and cooperative binding of some transcription factors 75 prevents these methods from accurately predicting binding of all transcription factors. A more re- 76 cent approach uses matrix completion to impute TF binding using a 3-mode tensor representing 22 77 genomic positions, cell types, and TF binding . This method doesn't rely on sequence specificity, 78 but can only predict TF binding in well-studied cell types with many ChIP-seq datasets. This 79 means one cannot use it to predict binding in a cell type where ChIP-seq is not possible, such as 80 limited clinical samples. 81 Identifying the best approach for predicting TF binding remains a challenge, because most 2 a100 75 50 25 0 RB1 TBP PML MAZ SIX5 TAF1 ATF1 TAF7 ATF7 BMI1 MXI1 ZZZ3 E2F1 MTA3 MTA1 EZH2 ZHX1 PHF8 HSF1 ZHX2 BRF1 BRF2 RFX5 PBX3 UBTF RNF2 CBX2 CBX3 CBX8 CBX5 BDP1 CHD1 CHD7 CHD4 CHD2 IKZF1 SMC3 MBD4 SIRT6 ZMIZ1 SIN3B SIN3A CREM − NONO ZFP36 EP300 KAT2B KAT2A SUZ12 H3F3A SAP30 TFDP1 FOXP2 NELFE ZBED1 CTBP2 GTF2B RAD21 NR3C1 NR1H2 RBBP5 MYBL2 HDAC2 HDAC6 HDAC1 HCFC1 CCNT2 BRCA1 CREB1 CEBPZ KDM4A KDM5A KDM5B CEBPD KDM1A SMAD2 RCOR1 NCOR1 TRIM22 TRIM28 HMGN3 WHSC1 ZNF143 ARID3A NANOG NFATC1 GTF2F1 GTF3C2 WRNIP1 SREBF2 SETDB1 TARDBP POLR3A POLR2A POLR3G CREBBP TBL1XR1 HA ZC3H11A CREB3L1 SUPT20H ZKSCAN1 NEUROD1 100 75 Peaks with sequence motif (%) Peaks 50 bioRxiv preprint 25 low. In contrast, most MAFKmatching peaks sequence both motif contain ofcompared its to the sequence TFs motif same with and TF, motif(c) show occupancy such central of as enrichment. 50% ATF3, orgreen), more. central and enrichment TFs where of 50%JASPAR the or (red), more motif TFs of is where peaksIndividual also less have the points: than sequence 50% outliers motif of (high beyonddian. motif peaks Box a occupancy, range: have blue). whisker. interquartile the range sequencedistribution (IQR). among motif Whisker: datasets most (low from extreme motif different valueChIP-seq occupancy, cell within peaks types quartile for and a TF replicates.Figure with Horizontal any line 1: JASPAR of sequence boxplot: motif from me- the TF's family. Boxplots show the 0 Central enrichment SP2 SP1 SP4 YY1 JUN SRF FOS MNT IRF4 IRF3 MYB MAX IRF2 SPI1 MYC TAL1 ATF3 ATF2 NFIC E2F4 ELF1 E2F6 KLF5 BATF PAX5 PAX8 ELK1 ELK4 TCF3 TCF7 EBF1 ZEB1 ETV6 ETS1 BCL3 USF1 NFE2 USF2 RFX1 NFYA PBX2 SOX6 NRF1 RELA JUND CTCF NFYB REST CUX1 MAFF EGR1 RXRA MAFK STAT1 STAT3 GFI1B MEIS2 GATA2 GATA1 GATA3 TCF12 NR2F2 FOSL2 FOSL1 FOXA1 FOXA2 CTCFL TEAD4 THAP1 HNF4A NR2C2 BACH1 GABPA MEF2A RUNX3 RUNX1 FOXM1 HNF4G MEF2C CEBPB SMAD1 ESRRA STAT5A ZNF384 ZNF217 ZNF274 ZNF263 TCF7L2 ZNF207 NFE2L2 ZBTB33 BCL11A ZBTB7A BCLAF1 POU5F1 POU2F2 SREBF1 BHLHE40 SMARCA4 SMARCB1 SMARCC1 doi: Most ChIP-seq peaks lack the TF's sequence motif. (a) Chromatin factors with ChIP-seq data in ENCODE dataset certified bypeerreview)istheauthor/funder.Allrightsreserved.Noreuseallowedwithoutpermission. https://doi.org/10.1101/168419 Motif occupancy No sequence motif 0% < Motif occupancy < 50% Motif occupancy ≥ 50% JASPAR MA0496.1 MAFK b d 0.04 Peaks with motif = 51569 2.0 No sequence motif 1.5 1.0 Bits 0.5 0.0 0% < Motif occupancy < 50% 19 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 0.03 of a TF's motif is lower for TFs with motif occupancy of less than 50% JASPAR MA0605.1 Atf3 Peaks with motif = 880 2.0 1.5 Motif occupancy ≥ 50% 1.0 Bits 0.5 0.0 0 20 40 60 80 1 2 3 4 5 6 7 8 Number of chromatin factors 0.02 ; this versionpostedMarch12,2019.
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]
  • Core Transcriptional Regulatory Circuitries in Cancer
    Oncogene (2020) 39:6633–6646 https://doi.org/10.1038/s41388-020-01459-w REVIEW ARTICLE Core transcriptional regulatory circuitries in cancer 1 1,2,3 1 2 1,4,5 Ye Chen ● Liang Xu ● Ruby Yu-Tong Lin ● Markus Müschen ● H. Phillip Koeffler Received: 14 June 2020 / Revised: 30 August 2020 / Accepted: 4 September 2020 / Published online: 17 September 2020 © The Author(s) 2020. This article is published with open access Abstract Transcription factors (TFs) coordinate the on-and-off states of gene expression typically in a combinatorial fashion. Studies from embryonic stem cells and other cell types have revealed that a clique of self-regulated core TFs control cell identity and cell state. These core TFs form interconnected feed-forward transcriptional loops to establish and reinforce the cell-type- specific gene-expression program; the ensemble of core TFs and their regulatory loops constitutes core transcriptional regulatory circuitry (CRC). Here, we summarize recent progress in computational reconstitution and biologic exploration of CRCs across various human malignancies, and consolidate the strategy and methodology for CRC discovery. We also discuss the genetic basis and therapeutic vulnerability of CRC, and highlight new frontiers and future efforts for the study of CRC in cancer. Knowledge of CRC in cancer is fundamental to understanding cancer-specific transcriptional addiction, and should provide important insight to both pathobiology and therapeutics. 1234567890();,: 1234567890();,: Introduction genes. Till now, one critical goal in biology remains to understand the composition and hierarchy of transcriptional Transcriptional regulation is one of the fundamental mole- regulatory network in each specified cell type/lineage.
    [Show full text]
  • Multiple Facets of Jund Gene Expression Are Atypical Among AP-1 Family Members
    Oncogene (2008) 27, 4757–4767 & 2008 Macmillan Publishers Limited All rights reserved 0950-9232/08 $30.00 www.nature.com/onc REVIEW Multiple facets of junD gene expression are atypical among AP-1 family members JM Hernandez1, DH Floyd2, KN Weilbaecher2, PL Green1,3 and K Boris-Lawrie1,3 1Department of Veterinary Biosciences and Center for Retrovirus Research, The Ohio State University, Columbus, OH, USA and 2Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St Louis, MO, USA and 3Department of Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA JunD is a versatile AP-1 transcription factor that can 2003; Milde-Langosch, 2005). The AP-1 component activate or repress a diverse collection of target genes. proteins are characterized structurally by their leucine- Precise control of junD expression and JunD protein– zipper dimerization motif and basic DNA-binding protein interactions modulate tumor angiogenesis, cellular domain. They can either activate or repress transcription differentiation, proliferation and apoptosis. Molecular and this versatile functional activity is dependent on the and clinical knowledge of two decades has revealed specific components of the dimeric complex and the that precise JunD activity is elaborated by interrelated cellular environment (Eferl and Wagner, 2003; Hess layers of constitutive transcriptional control, complex et al., 2004). AP-1 figures prominently in transcriptional post-transcriptional regulation and a collection of regulation of early response genes (reviewed by Jochum post-translational modifications and protein–protein et al., 2001; Mechta-Grigoriou et al., 2001; Eferl and interactions. The stakes are high, as inappropriate JunD Wagner, 2003).
    [Show full text]
  • FOXA2 Is a Sensitive and Specific Marker for Small Cell Neuroendocrine Carcinoma of the Prostate Jung Wook Park1, John K Lee2,3, Owen N Witte1,4,5 and Jiaoti Huang6
    Modern Pathology (2017) 30, 1262–1272 1262 © 2017 USCAP, Inc All rights reserved 0893-3952/17 $32.00 FOXA2 is a sensitive and specific marker for small cell neuroendocrine carcinoma of the prostate Jung Wook Park1, John K Lee2,3, Owen N Witte1,4,5 and Jiaoti Huang6 1Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California—Los Angeles, Los Angeles, CA, USA; 2Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California—Los Angeles, Los Angeles, CA, USA; 3Molecular Biology Institute, David Geffen School of Medicine, University of California— Los Angeles, Los Angeles, CA, USA; 4Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California—Los Angeles, Los Angeles, CA, USA; 5Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California—Los Angeles, Los Angeles, CA, USA and 6Department of Pathology, Duke University School of Medicine, Durham, NC, USA The median survival of patients with small cell neuroendocrine carcinoma is significantly shorter than that of patients with classic acinar-type adenocarcinoma. Small cell neuroendocrine carcinoma is traditionally diagnosed based on histologic features because expression of current immunohistochemical markers is inconsistent. This is a challenging diagnosis even for expert pathologists and particularly so for pathologists who do not specialize in prostate cancer. New biomarkers to aid in the diagnosis of small cell neuroendocrine carcinoma are therefore urgently needed. We discovered that FOXA2, a pioneer transcription factor, is frequently and specifically expressed in small cell neuroendocrine carcinoma compared with prostate adenocarcinoma from published mRNA-sequencing data of a wide range of human prostate cancers.
    [Show full text]
  • Charting Brachyury-Mediated Developmental Pathways During Early Mouse Embryogenesis
    Charting Brachyury-mediated developmental pathways during early mouse embryogenesis Macarena Lolasa,b, Pablo D. T. Valenzuelab, Robert Tjiana,c,1, and Zhe Liud,1 dJunior Fellow Program, aJanelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147; bFundación Ciencia para la Vida, Santiago 7780272, Chile; and cLi Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine Center of Excellence, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720 Contributed by Robert Tjian, February 11, 2014 (sent for review January 14, 2014) To gain insights into coordinated lineage-specification and mor- cells play diverse and indispensable roles in early mouse phogenetic processes during early embryogenesis, here we report development. a systematic identification of transcriptional programs mediated In mouse ES cell-based differentiation systems, Brachyury is by a key developmental regulator—Brachyury. High-resolution widely used as a mesoendoderm marker, and Brachyury-positive chromosomal localization mapping of Brachyury by ChIP sequenc- cells were able to differentiate into mesodermal and definitive ing and ChIP-exonuclease revealed distinct sequence signatures endodermal lineages, such as cardiomyocytes and hepatocytes enriched in Brachyury-bound enhancers. A combination of genome- (8–10). In a previous study, we found that depletion of a core wide in vitro and in vivo perturbation analysis and cross-species promoter factor, the TATA binding protein-associated factor 3, evolutionary comparison unveiled a detailed Brachyury-depen- in ES cells leads to significant up-regulation of Brachyury and dent gene-regulatory network that directly links the function of mesoderm lineages during ES cell differentiation (11). Here, we Brachyury to diverse developmental pathways and cellular house- systematically characterized the molecular function of Brachyury keeping programs.
    [Show full text]
  • Table S1 the Four Gene Sets Derived from Gene Expression Profiles of Escs and Differentiated Cells
    Table S1 The four gene sets derived from gene expression profiles of ESCs and differentiated cells Uniform High Uniform Low ES Up ES Down EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol 269261 Rpl12 11354 Abpa 68239 Krt42 15132 Hbb-bh1 67891 Rpl4 11537 Cfd 26380 Esrrb 15126 Hba-x 55949 Eef1b2 11698 Ambn 73703 Dppa2 15111 Hand2 18148 Npm1 11730 Ang3 67374 Jam2 65255 Asb4 67427 Rps20 11731 Ang2 22702 Zfp42 17292 Mesp1 15481 Hspa8 11807 Apoa2 58865 Tdh 19737 Rgs5 100041686 LOC100041686 11814 Apoc3 26388 Ifi202b 225518 Prdm6 11983 Atpif1 11945 Atp4b 11614 Nr0b1 20378 Frzb 19241 Tmsb4x 12007 Azgp1 76815 Calcoco2 12767 Cxcr4 20116 Rps8 12044 Bcl2a1a 219132 D14Ertd668e 103889 Hoxb2 20103 Rps5 12047 Bcl2a1d 381411 Gm1967 17701 Msx1 14694 Gnb2l1 12049 Bcl2l10 20899 Stra8 23796 Aplnr 19941 Rpl26 12096 Bglap1 78625 1700061G19Rik 12627 Cfc1 12070 Ngfrap1 12097 Bglap2 21816 Tgm1 12622 Cer1 19989 Rpl7 12267 C3ar1 67405 Nts 21385 Tbx2 19896 Rpl10a 12279 C9 435337 EG435337 56720 Tdo2 20044 Rps14 12391 Cav3 545913 Zscan4d 16869 Lhx1 19175 Psmb6 12409 Cbr2 244448 Triml1 22253 Unc5c 22627 Ywhae 12477 Ctla4 69134 2200001I15Rik 14174 Fgf3 19951 Rpl32 12523 Cd84 66065 Hsd17b14 16542 Kdr 66152 1110020P15Rik 12524 Cd86 81879 Tcfcp2l1 15122 Hba-a1 66489 Rpl35 12640 Cga 17907 Mylpf 15414 Hoxb6 15519 Hsp90aa1 12642 Ch25h 26424 Nr5a2 210530 Leprel1 66483 Rpl36al 12655 Chi3l3 83560 Tex14 12338 Capn6 27370 Rps26 12796 Camp 17450 Morc1 20671 Sox17 66576 Uqcrh 12869 Cox8b 79455 Pdcl2 20613 Snai1 22154 Tubb5 12959 Cryba4 231821 Centa1 17897
    [Show full text]
  • Harnessing Gene Expression Profiles for the Identification of Ex Vivo Drug
    cancers Article Harnessing Gene Expression Profiles for the Identification of Ex Vivo Drug Response Genes in Pediatric Acute Myeloid Leukemia David G.J. Cucchi 1 , Costa Bachas 1 , Marry M. van den Heuvel-Eibrink 2,3, Susan T.C.J.M. Arentsen-Peters 3, Zinia J. Kwidama 1, Gerrit J. Schuurhuis 1, Yehuda G. Assaraf 4, Valérie de Haas 3 , Gertjan J.L. Kaspers 3,5 and Jacqueline Cloos 1,* 1 Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; [email protected] (D.G.J.C.); [email protected] (C.B.); [email protected] (Z.J.K.); [email protected] (G.J.S.) 2 Department of Pediatric Oncology/Hematology, Erasmus MC–Sophia Children’s Hospital, 3015 CN Rotterdam, The Netherlands; [email protected] 3 Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; [email protected] (S.T.C.J.M.A.-P.); [email protected] (V.d.H.); [email protected] (G.J.L.K.) 4 The Fred Wyszkowski Cancer Research, Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel; [email protected] 5 Emma’s Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1081 HV Amsterdam, The Netherlands * Correspondence: [email protected] Received: 21 April 2020; Accepted: 12 May 2020; Published: 15 May 2020 Abstract: Novel treatment strategies are of paramount importance to improve clinical outcomes in pediatric AML. Since chemotherapy is likely to remain the cornerstone of curative treatment of AML, insights in the molecular mechanisms that determine its cytotoxic effects could aid further treatment optimization.
    [Show full text]
  • Reversible Human Immunodeficiency Virus Type-1 Latency in Primary Human Monocyte-Derived Macrophages Induced by Sustained M1
    www.nature.com/scientificreports OPEN Reversible Human Immunodefciency Virus Type-1 Latency in Primary Human Received: 11 January 2018 Accepted: 13 August 2018 Monocyte-Derived Macrophages Published: xx xx xxxx Induced by Sustained M1 Polarization Francesca Graziano1,4, Giulia Aimola1, Greta Forlani 2, Filippo Turrini 1, Roberto S. Accolla2, Elisa Vicenzi1 & Guido Poli1,3 We have reported that short-term stimulation of primary human monocyte-derived macrophages (MDM) with interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), i.e. M1 polarization, leads to a signifcant containment of virus replication. Here we show that M1-MDM restimulation with these cytokines 7 days after infection (M12 MDM) promoted an increased restriction of HIV-1 replication characterized by very low levels of virus production near to undetectable levels. In comparison to control and M1-MDM that were not restimulated, M12 MDM showed a stronger reduction of both total and integrated HIV DNA as well as of viral mRNA expression. M12 MDM were characterized by an upregulated expression of restriction factors acting at the level of reverse transcription (RT), including apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3A (APOBEC3A) and APOBEC3G, but not SAM domain and HD domain-containing protein 1 (SAMHD1). M12 MDM also showed an increased expression of Class II Transactivator (CIITA) and Tripartite Motif22 (TRIM22), two negative regulators of proviral transcription, whereas expression and phosphorylation of transcriptional inducers of HIV-1, such as nuclear factor kB (NF-kB) and signal transducer and activator of transcription 1 (STAT1), were not impaired in these cells. The almost quiescent state of the infection in M12 MDM was promptly reversed by coculture with mitogen-stimulated leukocytes or cell incubation with their fltered culture supernatant.
    [Show full text]
  • Impact of ZBTB7A Hypomethylation and Expression Patterns on Treatment Response to Hydroxyurea Vasiliki Chondrou1†, Eleana F
    Chondrou et al. Human Genomics (2018) 12:45 https://doi.org/10.1186/s40246-018-0177-z PRIMARY RESEARCH Open Access Impact of ZBTB7A hypomethylation and expression patterns on treatment response to hydroxyurea Vasiliki Chondrou1†, Eleana F. Stavrou1†, Georgios Markopoulos2, Alexandra Kouraklis-Symeonidis3, Vasilios Fotopoulos4, Argiris Symeonidis5, Efthymia Vlachaki6, Panagiota Chalkia7, George P. Patrinos8, Adamantia Papachatzopoulou9 and Argyro Sgourou1* Abstract Background: We aimed to clarify the emerging epigenetic landscape in a group of genes classified as “modifier genes” of the β-type globin genes (HBB cluster), known to operate in trans to accomplish the two natural developmental switches in globin expression, from embryonic to fetal during the first trimester of conception and from fetal to adult around the time of birth. The epigenetic alterations were determined in adult sickle cell anemia (SCA) homozygotes and SCA/β-thalassemia compound heterozygotes of Greek origin, who are under hydroxyurea (HU) treatment. Patients were distinguished in HU responders and HU non-responders (those not benefited from the HU) and both, and in vivo and in vitro approaches were implemented. Results: We examined the CpG islands’ DNA methylation profile of BCL11A, KLF1, MYB, MAP3K5, SIN3A, ZBTB7A,andGATA2, along with γ-globin and LRF/ZBTB7A expression levels. In vitro treatment of hematopoietic stem cells (HSCs) with HU induced a significant DNA hypomethylation pattern in ZBTB7A (p*, 0.04) and GATA2 (p*, 0.03) CpGs exclusively in the HU non-responders. Also, this group of patients exhibited significantly elevated baseline methylation patterns in ZBTB7A, before the HU treatment, compared to HU responders (p*, 0.019) and to control group of healthy individuals (p*, 0.021) , which resembles a potential epigenetic barrier for the γ-globin expression.
    [Show full text]
  • Genome-Wide Inference of Natural Selection on Human Transcription Factor Binding Sites
    ANALYSIS Genome-wide inference of natural selection on human transcription factor binding sites Leonardo Arbiza1, Ilan Gronau1, Bulent A Aksoy2, Melissa J Hubisz1, Brad Gulko3, Alon Keinan1–3 & Adam Siepel1–3 For decades, it has been hypothesized that gene regulation persistence in humans7,8. In addition, some genome-wide analyses has had a central role in human evolution, yet much remains have found bulk statistical evidence of natural selection in noncoding unknown about the genome-wide impact of regulatory regions near genes, presumably due to cis-regulatory elements9–12. mutations. Here we use whole-genome sequences and genome- Nevertheless, evidence in support of the overall prominence of wide chromatin immunoprecipitation and sequencing data to cis-regulatory mutations in evolutionary adaptation remains largely demonstrate that natural selection has profoundly influenced anecdotal and indirect, and there is continuing controversy about the human transcription factor binding sites since the divergence relative roles of regulatory and protein-coding sequences in evolu- of humans from chimpanzees 4–6 million years ago. Our tion8. Large-scale genomic studies of the evolution of transcription analysis uses a new probabilistic method, called INSIGHT, for factor binding sites have the potential to advance this debate, but a measuring the influence of selection on collections of short, major limitation of such studies so far has been a lack of precisely interspersed noncoding elements. We find that, on average, annotated binding sites across the genome. The analysis of con- transcription factor binding sites have experienced somewhat served noncoding sequences and/or promoter regions rather than weaker selection than protein-coding genes.
    [Show full text]
  • The New Therapeutic Strategies in Pediatric T-Cell Acute Lymphoblastic Leukemia
    International Journal of Molecular Sciences Review The New Therapeutic Strategies in Pediatric T-Cell Acute Lymphoblastic Leukemia Marta Weronika Lato 1 , Anna Przysucha 1, Sylwia Grosman 1, Joanna Zawitkowska 2 and Monika Lejman 3,* 1 Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; [email protected] (M.W.L.); [email protected] (A.P.); [email protected] (S.G.) 2 Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; [email protected] 3 Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland * Correspondence: [email protected] Abstract: Childhood acute lymphoblastic leukemia is a genetically heterogeneous cancer that ac- counts for 10–15% of T-cell acute lymphoblastic leukemia (T-ALL) cases. The T-ALL event-free survival rate (EFS) is 85%. The evaluation of structural and numerical chromosomal changes is important for a comprehensive biological characterization of T-ALL, but there are currently no ge- netic prognostic markers. Despite chemotherapy regimens, steroids, and allogeneic transplantation, relapse is the main problem in children with T-ALL. Due to the development of high-throughput molecular methods, the ability to define subgroups of T-ALL has significantly improved in the last few years. The profiling of the gene expression of T-ALL has led to the identification of T-ALL subgroups, and it is important in determining prognostic factors and choosing an appropriate treatment. Novel therapies targeting molecular aberrations offer promise in achieving better first remission with the Citation: Lato, M.W.; Przysucha, A.; hope of preventing relapse.
    [Show full text]
  • SOX4-Mediated Repression of Specific Trnas Inhibits Proliferation of Human Glioblastoma Cells
    SOX4-mediated repression of specific tRNAs inhibits proliferation of human glioblastoma cells Jianjing Yanga,b,c, Derek K. Smithc,d, Haoqi Nia,b,c,KeWua,b, Dongdong Huanga,b, Sishi Pana,b,c, Adwait A. Sathee, Yu Tangc,d, Meng-Lu Liuc,d, Chao Xinge,f,g, Chun-Li Zhangc,d,1, and Qichuan Zhugea,b,1 aDepartment of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China 325000; bZhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China 325000; cDepartment of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390; dHamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390; eMcDermott Center of Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390; fDepartment of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390; and gDepartment of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390 Edited by S. Altman, Yale University, New Haven, CT, and approved February 5, 2020 (received for review November 15, 2019) Transfer RNAs (tRNAs) are products of RNA polymerase III (Pol III) indicates that tRNA expression may also be under cell state- and essential for mRNA translation and ultimately cell growth and dependent regulations (12–16). proliferation. Whether and how individual tRNA genes are specif- In this study, we performed a systematic analysis on how ically regulated is not clear. Here, we report that SOX4, a well- NGN2/SOX4-mediated cell-fate reprogramming leads to cell known Pol II-dependent transcription factor that is critical for neuro- cycle exit of human glioblastoma cells.
    [Show full text]