P68/Ddx5 RNA Helicase Interacts and Co-Localizes in Vivo with the De Novo DNA Methyltransferases Dnmt3a1 and Dnmt3a2 Anastasia Mpakali1,2#, Andriana G

Total Page:16

File Type:pdf, Size:1020Kb

P68/Ddx5 RNA Helicase Interacts and Co-Localizes in Vivo with the De Novo DNA Methyltransferases Dnmt3a1 and Dnmt3a2 Anastasia Mpakali1,2#, Andriana G ics om & B te i ro o P in f f o o r l m a Journal of a n t r i Mpakali et al., J Proteomics Bioinform 2012, 5:1 c u s o J DOI: 10.4172/jpb.1000207 ISSN: 0974-276X Proteomics & Bioinformatics Research Article Article OpenOpen Access Access P68/Ddx5 RNA Helicase Interacts and Co-Localizes In vivo with the De Novo DNA Methyltransferases Dnmt3a1 and Dnmt3a2 Anastasia Mpakali1,2#, Andriana G. Kotini1,2#, Magda Spella3, Marina Kouyialis2, Angelliki Tserga2, Leonidas Fragkos-Livanios4, Martina Samiotaki4 and Theodora Agalioti2,3* 1National and Kapodistrian University of Athens-Medical School 2BSRC-Alexander Fleming-Institute of Molecular Biology and Genetics 3Laboratory for Molecular Respiratory Carcinogenesis- Department of Physiology, Faculty of Medicine, University of Patras 4BSRC-Alexander Fleming-Institute of Molecular Oncology #These authors contributed equally Abstract The 5-methyl cytosine (5meC) genomic methylation patterns play crucial roles in mammalian development and are altered in cancer. The enzymes that create, maintain and modify the DNA methylation patterns are the DNA methyltransferases (Dnmts) which are all encoded by essential genes. The de novo Dnmts -Dnmt3a and Dnmt3b- establish the DNA methylation patterns early in mammalian development by introducing DNA methylation marks where no previous methylation exists. These enzymes do not exhibit affinity for specific DNA sequences, thus their recruitment to specific DNA loci and their activities must be tightly regulated. In particular, Dnmt3a2 –one of the two protein isoforms produced by the Dnmt3a locus- is the most abundant DNA methyltransferase in mouse Embryonic Stem Cells. To identify Dnmt3a (and DNA methylation) regulators we have searched for Dnmt3a2 interacting proteins in mESCs by pull down and Mass Spectrometry. The DEAD box p68/Ddx5 RNA helicase was identified to directly interact with Dnmt3a1 and Dnmt3a2 in vitro and in vivo. We have created a mutant Ddx5 (Ddx5MUT) protein exhibiting an altered nuclear localization pattern compared to the wt protein. Both wt and mutant Ddx5 interact directly in vitro and co localize in vivo with Dnmt3a proteins. Our data suggest that the Dnmt3a/Ddx5 interaction might be significant for modulating the DNA methylation/demethylation dynamics in vivo. Introduction P68/Ddx5 is a prototypical member of the DEAD box family of RNA helicases exhibiting ATPase and RNA unwinding activities. DNA methylation in mammals is associated with important The DEAD box family is named after the amino acid sequence of its developmental phenomena such as genomic Imprinting and conserved Motif II (also known as the Walker B motif) containing the X-inactivation and is inversely correlated with transcription initiation [1]. The DNA methylation patterns, thought to characterize each cell amino acids asp-glu-ala-asp (D-E-A-D). Ddx5 is a multifunctional type as a molecular fingerprint, are established early in mammalian protein involved in transcription regulation, mRNA splicing, miRNA development and are altered in cancer [2-5]. The mouse DNA biogenesis and in DNA damage repair pathways [13]. In particular, methyltransferases (Dnmts), the enzymes that create, maintain and in DNA repair, Ddx5 is associated with Thymidine DNA glycosylase modify the DNA methylation patterns are all encoded by essential (TDG), a protein which is essential for active DNA demethylation [14]. genes. These enzymes are categorized according to their ability to However, the Ddx5 roles in this context are not well understood. introduce new DNA methylation marks into genomic regions where New, exciting findings point towards the notion that the DNA no previous DNA methylation exists (de novo Dnmts) or to copy methylation patterns, thought to characterize each cell type as a pre-existing methylation into newly synthesized DNA strands in the context of the genomic CpG nucleotides (maintenance enzyme, molecular fingerprint are dynamically built up and erased during Dnmt1) [6-9]. The de novo Dnmts belong to the Dnmt3 family which development and in adult tissues [15-17]. Currently, our understanding contains three members, namely Dnmt3a, Dnmt3b and Dnmt3L. of the DNA methylation/demethylation dynamics involves the actions Dnmt3a and Dnmt3b are active enzymes while Dnmt3L is an auxiliary of several groups of enzymes such as a) the DNA methyltransferases molecule enhancing the enzymatic activity of Dnmt3a/3b proteins (Dnmts) which introduce and maintain the 5-methylcytosine (5meC), [10]. The Dnmt3a locus (together with Dnmt3L) is responsible for b) the ten-eleven translocation (TET) family of enzymes that modifies establishing the differential DNA methylation marks associated with genomic Imprinting, which results into the “parent of origin”- specific monoallelic expression of ~100 autosomal genes [11]. Additionally, *Corresponding author: Theodora Agalioti, Laboratory for Molecular Respiratory DNMT3A is associated with cancer pathogenesis because it is found Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Basic Biomedical Sciences Research Building, 2nd Fl, University Campus, mutated in 20% of acute myeloid leukemia patients [12]. 1 Asklepiou Str, GR-26504 Patras-Greece, Tel: +30 2610 969116; Fax: +30 2610 997215; E-mail: [email protected], [email protected] The Dnmt3a genomic locus produces two proteins differing in that Received December 04, 2011; Accepted January 10, 2012; Published January a 219aa unique N-terminal “tail” lacking from the Dnmt3a2 is present 16, 2012 in the longer isoform Dnmt3a1. Dnmt3a1 and Dnmt3a2 proteins are Citation: Mpakali A, Kotini AG, Spella M, Kouyialis M, Tserga A, et al. (2012) P68/ expressed in mouse Embryonic Stem Cells (mESCs) and in early stages Ddx5 RNA Helicase Interacts and Co-Localizes In vivo with the DNA of mouse development which are thought to be enriched in Stem and Methyltransferases Dnmt3a1 and Dnmt3a2. J Proteomics Bioinform 5: 007-014. precursor cells. Dnmt3a1 is expressed in low levels in adult somatic doi:10.4172/jpb.1000207 cells. In contrast, the Dnmt3a2 mRNA and protein expression ceases to Copyright: © 2012 Mpakali A, et al. This is an open-access article distributed under undetectable levels in later stages of development, in adult somatic cells the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and and upon mESCs differentiation [6]. source are credited. J Proteomics Bioinform ISSN:0974-276X JPB, an open access journal Volume 5(1) : 007-014 (2012) - 007 Citation: Mpakali A, Kotini AG, Spella M, Kouyialis M, Tserga A, et al. (2012) P68/Ddx5 RNA Helicase Interacts and Co-Localizes In vivo with the DNA Methyltransferases Dnmt3a1 and Dnmt3a2. J Proteomics Bioinform 5: 007-014. doi:10.4172/jpb.1000207 5meC by successive hydroxylation and oxidation towards 5-hydroxy- His-Ddx5 or with His-Ddx5MUT proteins. The beads were washed twice methyl cytosine (5hmC), c) the activation induced cytidine deaminases with PBS+0.01% Tween and five times with PBS+0.01% Tween plus 0.3 (AID and Apobec 1-3) which are thought to deaminate (5meC or M NaCl. The beads were subsequently boiled in SDS PAGE buffer and 5hmC) and d) the DNA glycosylases of the Base Excision Repair (BER) analyzed in Western blots. pathway initiating the replacement of modified cytosines (5meC or Fluorescence and confocal microscopy 5hmC) with unmodified ones thus, effecting DNA demethylation. After 24h the cover slips bearing the transfected cells were washed To get insights on how the DNA methylation/demethylation twice with PBS and the cells were fixed in 4% PFA for 10 min at RT. The dynamics are regulated we sought to identify Dnmt3a2 interacting cells were washed twice for 5min with PBS. Excess fluid was removed proteins in Mouse Embryonic Stem Cells (mESCs). We have identified and the cover slips were placed “face down” on a drop of ProLong Gold that the p68/Ddx5 DEAD box RNA helicase is specifically pulled down anti-fade reagent with DAPI (Invitrogen) on objective slides, so as the from mESCs nuclear extract preparations by a GST-tagged Dnmt3a2 fixed cells to be immersed into the DAPI containing solution. Excess bait protein. We show that the interaction between the two proteins fluid was removed and the preparations were left to dry for 30 min is direct. To explore the functional significance of this interaction in at RT in the dark. Direct fluorescence (figure 3 of this manuscript) vivo, we have created a Ddx5 mutation that is expected to be an active was monitored on the Observer D1 fluorescence microscope (Zeiss). ATPase with a compromised Helicase activity. We have expressed Areas of interest were scanned with x63 objective lens and images fluorescently tagged (GFP and RFP) versions of Dnmt3a1/3a2 and wt were captured by the AxioVision Rel. 4.8.2 program (Zeiss). The co- and mutant Ddx5 and examined their sub-cellular localization. We localization studies of the co-transfected protein constructs were find that unlike wt Ddx5, the mutant protein does not form nuclear conducted with the use of a Leica TCS SP5 with the dual (Tandem) speckles. Both wt and mutant Ddx5 proteins co-localize better with Scanner (Leica). Green and Red direct fluorescence were excited by Dnmt3a2 and to a lesser extent with Dnmt3a1 in the cell nucleus. laser beams at 488nm and 555nm respectively. Emissions were then sequentially acquired. Areas of interest were scanned with a x40 oil Materials and Methods immersion objective lens. Cell lines, plasmids and transfections Silver staining HEK 293T were maintained in ES medium [DMEM supplemented The following modified silver staining protocol which is compatible with 10% Fetal Bovine Serum (PAN Biotech), 50U/ml Penicillin/ with the LC-MS/MS analysis, was used for the SDS PAGE gel treatment. Streptomycin, 2mM L-Glutamine and 0.1 mM β-Merkaptoethanol. Briefly, the gels were immersed in a solution of 50% Methanol, 5% The cells were grown on round cover slips placed in tissue culture Acetic Acid for 20 min, followed by a 10 min wash in 50% methanol dishes and coated with 0.1% gelatin.
Recommended publications
  • Functional Control of HIV-1 Post-Transcriptional Gene Expression by Host Cell Factors
    Functional control of HIV-1 post-transcriptional gene expression by host cell factors DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Amit Sharma, B.Tech. Graduate Program in Molecular Genetics The Ohio State University 2012 Dissertation Committee Dr. Kathleen Boris-Lawrie, Advisor Dr. Anita Hopper Dr. Karin Musier-Forsyth Dr. Stephen Osmani Copyright by Amit Sharma 2012 Abstract Retroviruses are etiological agents of several human and animal immunosuppressive disorders. They are associated with certain types of cancer and are useful tools for gene transfer applications. All retroviruses encode a single primary transcript that encodes a complex proteome. The RNA genome is reverse transcribed into DNA, integrated into the host genome, and uses host cell factors to transcribe, process and traffic transcripts that encode viral proteins and act as virion precursor RNA, which is packaged into the progeny virions. The functionality of retroviral RNA is governed by ribonucleoprotein (RNP) complexes formed by host RNA helicases and other RNA- binding proteins. The 5’ leader of retroviral RNA undergoes alternative inter- and intra- molecular RNA-RNA and RNA-protein interactions to complete multiple steps of the viral life cycle. Retroviruses do not encode any RNA helicases and are dependent on host enzymes and RNA chaperones. Several members of the host RNA helicase superfamily are necessary for progressive steps during the retroviral replication. RNA helicase A (RHA) interacts with the redundant structural elements in the 5’ untranslated region (UTR) of retroviral and selected cellular mRNAs and this interaction is necessary to facilitate polyribosome formation and productive protein synthesis.
    [Show full text]
  • The RNA Helicase a in Malignant Transformation
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 19 The RNA helicase A in malignant transformation Marco Fidaleo1,2, Elisa De Paola1,2 and Maria Paola Paronetto1,2 1 Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy 2 Laboratory of Cellular and Molecular Neurobiology, CERC, Fondazione Santa Lucia, Rome, Italy Correspondence to: Maria Paola Paronetto, email: [email protected] Keywords: RHA helicase, genomic stability, cancer Received: October 02, 2015 Accepted: January 29, 2016 Published: February 14, 2016 ABSTRACT The RNA helicase A (RHA) is involved in several steps of RNA metabolism, such as RNA processing, cellular transit of viral molecules, ribosome assembly, regulation of transcription and translation of specific mRNAs. RHA is a multifunctional protein whose roles depend on the specific interaction with different molecular partners, which have been extensively characterized in physiological situations. More recently, the functional implication of RHA in human cancer has emerged. Interestingly, RHA was shown to cooperate with both tumor suppressors and oncoproteins in different tumours, indicating that its specific role in cancer is strongly influenced by the cellular context. For instance, silencing of RHA and/or disruption of its interaction with the oncoprotein EWS-FLI1 rendered Ewing sarcoma cells more sensitive to genotoxic stresses and affected tumor growth and maintenance, suggesting possible therapeutic implications. Herein, we review the recent advances in the cellular functions of RHA and discuss its implication in oncogenesis, providing a perspective for future studies and potential translational opportunities in human cancer. INTRODUCTION females, which contain two X chromosomes [8]. The C. elegans RHA-1 displays about 60% of similarity with both RNA helicase A (RHA) is a DNA/RNA helicase human RHA and Drosophila MLE and is involved in gene involved in all the essential steps of RNA metabolism, silencing.
    [Show full text]
  • Integrating Many Co-Splicing Networks to Reconstruct Splicing Regulatory Modules
    Dai et al. BMC Systems Biology 2012, 6(Suppl 1):S17 http://www.biomedcentral.com/1752-0509/6/S1/S17 RESEARCH Open Access Integrating many co-splicing networks to reconstruct splicing regulatory modules Chao Dai1,2†, Wenyuan Li2†, Juan Liu1, Xianghong Jasmine Zhou2* From The 5th IEEE International Conference on Computational Systems Biology (ISB 2011) Zhuhai, China. 02-04 September 2011 Abstract Background: Alternative splicing is a ubiquitous gene regulatory mechanism that dramatically increases the complexity of the proteome. However, the mechanism for regulating alternative splicing is poorly understood, and study of coordinated splicing regulation has been limited to individual cases. To study genome-wide splicing regulation, we integrate many human RNA-seq datasets to identify splicing module, which we define as a set of cassette exons co-regulated by the same splicing factors. Results: We have designed a tensor-based approach to identify co-splicing clusters that appear frequently across multiple conditions, thus very likely to represent splicing modules - a unit in the splicing regulatory network. In particular, we model each RNA-seq dataset as a co-splicing network, where the nodes represent exons and the edges are weighted by the correlations between exon inclusion rate profiles. We apply our tensor-based method to the 38 co-splicing networks derived from human RNA-seq datasets and indentify an atlas of frequent co-splicing clusters. We demonstrate that these identified clusters represent potential splicing modules by validating against four biological knowledge databases. The likelihood that a frequent co-splicing cluster is biologically meaningful increases with its recurrence across multiple datasets, highlighting the importance of the integrative approach.
    [Show full text]
  • The BRCA1-Associated Protein BACH1 Is a DNA Helicase Targeted by Clinically Relevant Inactivating Mutations
    The BRCA1-associated protein BACH1 is a DNA helicase targeted by clinically relevant inactivating mutations Sharon Cantor*†, Ronny Drapkin†‡, Fan Zhang‡, Yafang Lin‡, Juliana Han‡, Sushmita Pamidi‡, and David M. Livingston‡§ ‡The Dana–Farber Cancer Institute, Harvard Medical School, Boston, MA 02115; and *Department of Cancer Biology, University of Massachusetts Medical School, Lazare Research Building, 364 Plantation Street, Worcester, MA 01605 Contributed by David M. Livingston, December 31, 2003 BACH1 is a nuclear protein that directly interacts with the highly pose patients to tumor development and are the products of conserved, C-terminal BRCT repeats of the tumor suppressor, mutant helicase encoding genes (11). In addition, mutations in BRCA1. Mutations within the BRCT repeats disrupt the interaction two helicases, XPB and XPD, have been linked to xeroderma between BRCA1 and BACH1, lead to defects in DNA repair, and pigmentosum, Cockayne syndrome, and trichothiodystrophy result in breast and ovarian cancer. BACH1 is necessary for efficient (11); also, certain polymorphisms in XPD are associated with an double-strand break repair in a manner that depends on its increased risk of basal cell carcinoma and melanoma (12). association with BRCA1. Moreover, some women with early-onset Previously, we detected a potential association between the breast cancer and no abnormalities in either BRCA1 or BRCA2 carry presence of certain germline BACH1 sequence changes and germline BACH1 coding sequence changes, suggesting that abnor- breast cancer development (6). Two independent germline mal BACH1 function contributes to tumor induction. Here, we BACH1 alterations were detected among a cohort of 65 women show that BACH1 is both a DNA-dependent ATPase and a 5؅-to-3؅ with early-onset breast cancer.
    [Show full text]
  • Rna Helicase Ddx5 Regulates Platelet Derived Growth Factor Receptorand Androgen Receptor (Ar) Expression in Breast Cancer
    Georgia State University ScholarWorks @ Georgia State University Biology Dissertations Department of Biology 5-4-2020 RNA HELICASE DDX5 REGULATES PLATELET DERIVED GROWTH FACTOR RECEPTORAND ANDROGEN RECEPTOR (AR) EXPRESSION IN BREAST CANCER Neha Panchbhai Follow this and additional works at: https://scholarworks.gsu.edu/biology_diss Recommended Citation Panchbhai, Neha, "RNA HELICASE DDX5 REGULATES PLATELET DERIVED GROWTH FACTOR RECEPTORAND ANDROGEN RECEPTOR (AR) EXPRESSION IN BREAST CANCER." Dissertation, Georgia State University, 2020. https://scholarworks.gsu.edu/biology_diss/238 This Dissertation is brought to you for free and open access by the Department of Biology at ScholarWorks @ Georgia State University. It has been accepted for inclusion in Biology Dissertations by an authorized administrator of ScholarWorks @ Georgia State University. For more information, please contact [email protected]. RNA HELICASE DDX5 REGULATES PLATELET DERIVED GROWTH FACTOR RECEPTOR β AND ANDROGEN RECEPTOR (AR) EXPRESSION IN BREAST CANCER by NEHA ARUN PANCHBHAI Under the Direction of Zhi-Ren Liu, PhD ABSTRACT P68 RNA helicase, a prototypical member of the DEAD box family of RNA helicases is important in many biological processes, including early organ development. However, its aberrant expression contributes to tumor development and progression. In this study, we show that p68 upregulates the transcriptional expression of the growth factor receptor, PDGFRβ P68 knockdown in MDAMB231 and BT549 breast cancer cells significantly decreases mRNA and protein levels of PDGFRβ and EMT markers, resulting in decreased migration. We have previously shown that PDGF-BB induces p68 phosphorylation, resulting in EMT via nuclear translocation of β -catenin. Here, we show that p68 promotes migration in response to PDGF-BB stimulation via upregulation of PDGFRβ in breast cancer cells, suggesting that PDGFRβ is in turn regulated by p68 to maintain a positive feedback loop.
    [Show full text]
  • Supplementary Data
    Supplemental Material Materials and Methods Immunohistochemistry Primary antibodies used for validation studies include: mouse anti-desmoglein-3 (Cat. # 32-6300, Invitrogen, CA, USA; 1:25), rabbit anti-cytokeratin 4 (Cat. # ab11215, Abcam, Cambridge, MA, USA; 1:100), mouse anti-cytokeratin 16 (Cat. # ab8741, Abcam; 1:25), rabbit anti-desmoplakin antibody (Cat. # ab14418, Abcam; 1:200), mouse anti-vimentin (Cat. # M7020, Dako, Carpinteria, CA, USA; 1:100). Secondary antibodies conjugated with biotin (Vector, Burlingame, CA, USA) were used, diluted to 1:400. Tissues slides containing archival FFPE sections, or tissue micro arrays (TMA) consisting of 508 HNSCC and controls, were dewaxed in SafeClear II (Fisher Scientific, Pittsburgh, PA, USA) hydrated through graded alcohols, immersed in 3% hydrogen peroxide in PBS for 30 min to quench the endogenous peroxidase, and processed for antigen retrieval and immunostaining with the appropriate primary antibodies and biotinylated secondary antibodies as described (1), followed by the avidin-biotin complex method (Vector Stain Elite, ABC kit; Vector). Slides were washed and developed in 3,3'- diaminobenzidine (Sigma FASTDAB tablet; Sigma Chemical) under microscopic control, and counterstained with Mayer's hematoxylin. For each stained TMA the number of positive cells in each core was visually evaluated and the results expressed as a percentage of stained cells/ total number of cells. According to their immunoreactivity the tissues array cores were divided according to tumor differentiation, where the percentage of stained cells in the three tumor classes were scored as more than 5% and less than 25% of the cells stained, 26 to 50%, 51 to 75% or, 75 to 100%.
    [Show full text]
  • Osterix Represses Adipogenesis by Negatively Regulating Pparγ
    www.nature.com/scientificreports OPEN Osterix represses adipogenesis by negatively regulating PPARγ transcriptional activity Received: 28 June 2016 Younho Han1, Chae Yul Kim1, Heesun Cheong2 & Kwang Youl Lee1 Accepted: 03 October 2016 Osterix is a novel bone-related transcription factor involved in osteoblast differentiation, and bone Published: 18 October 2016 maturation. Because a reciprocal relationship exists between adipocyte and osteoblast differentiation of bone marrow derived mesenchymal stem cells, we hypothesized that Osterix might have a role in adipogenesis. Ablation of Osterix enhanced adipogenesis in 3T3-L1 cells, whereas overexpression suppressed this process and inhibited the expression of adipogenic markers including CCAAT/enhancer- binding protein alpha (C/EBPα) and peroxisome proliferator-activated receptor gamma (PPARγ). Further studies indicated that Osterix significantly decreased PPARγ-induced transcriptional activity. Using co-immunoprecipitation and GST-pull down analysis, we found that Osterix directly interacts with PPARγ. The ligand-binding domain (LBD) of PPARγ was responsible for this interaction, which was followed by repression of PPARγ-induced transcriptional activity, even in the presence of rosiglitazone. Taken together, we identified the Osterix has an important regulatory role on PPARγ activity, which contributed to the mechanism of adipogenesis. Obesity, characterized by excessive fat deposition due to an energy imbalance between energy intake and expend- iture, is a prevalent nutritional disorder, which tends to increase the risk of cardiovascular diseases, diabetes, and several types of cancer1,2. Obesity is a worldwide epidemic, with the prevalence of this condition steadily rising3. Therefore, there is a major need for therapeutic anti-obesity products that are proven to be safe and effective.
    [Show full text]
  • The Most Important Transcriptional Factors of Osteoblastogenesis
    ADVANCES IN CELL BIOLOGY VOL. 2, ISSUE 1/2010 (17–28) THE MOST IMPORTANT TRANSCRIPTIONAL FACTORS OF OSTEOBLASTOGENESIS Malgorzata WITKOWSKA-ZIMNY, Edyta WROBEL, Jacek PRZYBYLSKI Department of Biophysics and Human Physiology, Medical University of Warsaw DOI: 10.2478/v10052-010-0002-x Summary: One of the key issues of organogenesis is the understanding of mechanisms underlying the differentiation of progenitor cells into more specialized cells of individual tissues. Recent transcriptomic and proteomic approaches of molecular biology have led to the identification of several factors and mechanisms regulating morphogenesis at the genetic level which affect the function of already differentiated cells. In the last few years, several reports about osteoblastogenesis have been published. This review presents recent findings on the role of the most important transcription factors supporting bone formation. Key words: osteoblasts, transcriptional factors, Runx2, Osterix, osteoblastogenesis INTRODUCTION The specific structure of bone, its function and metabolism are a result of the processes of bone formation, resorption, mineral homeostasis and bone regeneration. These processes are based on the presence of specialized bone tissue cell types: osteoblasts, osteocytes and osteoclasts [22]. The structural and metabolic activity of bone is compromised by an imbalance between the rate of bone resorption and formation, which depends on osteoclasts and osteoblasts, respectively. In addition to the cellular elements, bone tissue is composed of extracellular matrix, which consists of organic components, including type I collagen (90%), osteocalcin, osteopontin, bone sialoprotein, osteonectin, proteoglycans and inorganic components – calcium phosphate salts in the form of hydroxyapatite [22]. Osteoblasts are bone-forming cells that synthesize and mineralize the extracellular matrix.
    [Show full text]
  • Goat Anti-DHX9 / RHA Antibody Peptide-Affinity Purified Goat Antibody Catalog # Af1317a
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 Goat Anti-DHX9 / RHA Antibody Peptide-affinity purified goat antibody Catalog # AF1317a Specification Goat Anti-DHX9 / RHA Antibody - Product Information Application WB Primary Accession Q08211 Other Accession NP_001348, 1660, 13211 (mouse), 304859 (rat) Reactivity Human Predicted Mouse, Rat, Cow Host Goat Clonality Polyclonal Concentration 100ug/200ul Isotype IgG AF1317a (1 µg/ml) staining of nuclear HeLa Calculated MW 140958 lysate (35 µg protein in RIPA buffer). Primary incubation was 1 hour. Detected by chemiluminescence. Goat Anti-DHX9 / RHA Antibody - Additional Information Goat Anti-DHX9 / RHA Antibody - Gene ID 1660 Background Other Names This gene encodes a member of the ATP-dependent RNA helicase A, RHA, DEAH-containing family of RNA helicases. The 3.6.4.13, DEAH box protein 9, Leukophysin, encoded protein is an enzyme that catalyzes LKP, Nuclear DNA helicase II, NDH II, DHX9, the ATP-dependent unwinding of DDX9, LKP, NDH2 double-stranded RNA and DNA-RNA complexes. This protein localizes to both the nucleus and Format the cytoplasm and functions as a 0.5 mg IgG/ml in Tris saline (20mM Tris transcriptional regulator. This protein may also pH7.3, 150mM NaCl), 0.02% sodium azide, be involved in the expression and nuclear with 0.5% bovine serum albumin export of retroviral RNAs. Alternate splicing Storage results in multiple transcript variants. Maintain refrigerated at 2-8°C for up to 6 Pseudogenes of this gene are found on months. For long term storage store at chromosomes 11 and 13.
    [Show full text]
  • Lncrna AK023948 Is a Positive Regulator of AKT
    ARTICLE Received 16 May 2016 | Accepted 28 Dec 2016 | Published 8 Feb 2017 DOI: 10.1038/ncomms14422 OPEN LncRNA AK023948 is a positive regulator of AKT Pratirodh Koirala1,2, Jianguo Huang1,2, Tsui-Ting Ho1,3,4, Fangting Wu5, Xianfeng Ding1,6 & Yin-Yuan Mo1,3 Despite the overwhelming number of human long non-coding RNAs (lncRNAs) reported so far, little is known about their physiological functions for the majority of them. The present study uses a CRISPR/Cas9-based synergistic activation mediator (SAM) system to identify potential lncRNAs capable of regulating AKT activity. Among lncRNAs identified from this screen, we demonstrate that AK023948 is a positive regulator for AKT. Knockout of AK023948 suppresses, whereas rescue with AK023948 restores the AKT activity. Mechanistically, AK023948 functionally interacts with DHX9 and p85. Importantly, AK023948 is required for the interaction between DHX9 and p85 to hence the p85 stability and promote AKT activity. Finally, AK023948 is upregulated in breast cancer; interrogation of TCGA data set indicates that upregulation of DHX9 in breast cancer is associated with poor survival. Together, this study demonstrates two previously uncharacterized factors AK023948 and DHX9 as important players in the AKT pathway, and that their upregulation may contribute to breast tumour progression. 1 Cancer Institute, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA. 2 Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA. 3 Department of Pharmacology/Toxicology, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA. 4 Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA. 5 System Biosciences, Mountain View, California 94041, USA.
    [Show full text]
  • Functional Characterization of the Dexh-Box Helicase DHX9
    Functional Characterization of the DExH-Box Helicase DHX9 Teresa Lee Department of Biochemistry McGill University Montréal, Québec, Canada October 2016 A thesis submitted to McGill University in partial fulfillment of the requirement of the degree of Doctor of Philosophy © Teresa Lee, October 2016 TABLE OF CONTENTS LIST OF FIGURES ...................................................................................................................... 6 LIST OF TABLES ........................................................................................................................ 8 ABSTRACT ................................................................................................................................... 9 RESUMÉ ..................................................................................................................................... 11 PREFACE .................................................................................................................................... 13 CONTRIBUTION OF AUTHORS ........................................................................................... 14 ORIGINAL CONTRIBUTION TO KNOWLEDGE .............................................................. 15 ACKNOWLEDGEMENTS ....................................................................................................... 17 LIST OF ABBREVIATIONS .................................................................................................... 18 CHAPTER 1: GENERAL INTRODUCTION ........................................................................
    [Show full text]
  • Structural Analysis of SARS-Cov-2 and Predictions of the Human Interactome
    A. Vandelli et al. Structure and interactions of SARS-CoV-2 Structural analysis of SARS-CoV-2 and predictions of the human interactome Andrea Vandelli1,2, Michele Monti1,3, Edoardo Milanetti4,5, Jakob Rupert 6, Elsa Zacco3, Elias Bechara1,3, Riccardo Delli Ponti 7,* and Gian Gaetano Tartaglia 1,3,6,8,* 1 Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain and Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain 2 Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain 3 Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy. 4 Department of Physics, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy 5 Center for Life Nanoscience, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy 6 Department of Biology ‘Charles Darwin’, Sapienza University of Rome, P.le A. Moro 5, Rome 00185, Italy 7 School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore 8 Institucio Catalana de Recerca i Estudis Avançats (ICREA), 23 Passeig Lluis Companys, 08010 Barcelona, Spain *to whom correspondence should be addressed to: [email protected] (RDP) and [email protected] or [email protected] (GGT) ABSTRACT Specific elements of viral genomes regulate interactions within host cells. Here, we calculated the secondary structure content of >2500 coronaviruses and computed >100000 human protein interactions with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We found that the 3’ and 5’ are the most structured elements in the viral genome and the 5’ has the strongest propensity to associate with human proteins.
    [Show full text]