MXD1 Localizes in the Nucleolus, Binds UBF and Impairs Rrna Synthesis

Total Page:16

File Type:pdf, Size:1020Kb

MXD1 Localizes in the Nucleolus, Binds UBF and Impairs Rrna Synthesis www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 43 Research Paper MXD1 localizes in the nucleolus, binds UBF and impairs rRNA synthesis Maria del Carmen Lafita-Navarro1,3, Rosa Blanco1, Jorge Mata-Garrido2, Judit Liaño-Pons1, Olga Tapia2, Lucía García-Gutiérrez1, Eva García-Alegría1,4, María T. Berciano2, Miguel Lafarga2, Javier León1 1Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria, and Department of Molecular Biology, University of Cantabria, Santander, Spain 2Department of Anatomy and Cell Biology and Centro de Investigación en Red sobre Enfermedades Neurodegenerativas (CIBERNED), University of Cantabria-IDIVAL, Santander, Spain 3Present address: Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas, USA 4Present address: Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom Correspondence to: Javier León, email: [email protected] Keywords: MXD1, UBF, nucleolus, pre-rRNA, transcription regulation Received: June 20, 2016 Accepted: August 26, 2016 Published: August 31, 2016 ABSTRACT MXD1 is a protein that interacts with MAX, to form a repressive transcription factor. MXD1-MAX binds E-boxes. MXD1-MAX antagonizes the transcriptional activity of the MYC oncoprotein in most models. It has been reported that MYC overexpression leads to augmented RNA synthesis and ribosome biogenesis, which is a relevant activity in MYC-mediated tumorigenesis. Here we describe that MXD1, but not MYC or MNT, localizes to the nucleolus in a wide array of cell lines derived from different tissues (carcinoma, leukemia) as well as in embryonic stem cells. MXD1 also localizes in the nucleolus of primary tissue cells as neurons and Sertoli cells. The nucleolar localization of MXD1 was confirmed by co-localization with UBF. Co-immunoprecipitation experiments showed that MXD1 interacted with UBF and proximity ligase assays revealed that this interaction takes place in the nucleolus. Furthermore, chromatin immunoprecipitation assays showed that MXD1 was bound in the transcribed rDNA chromatin, where it co-localizes with UBF, but also in the ribosomal intergenic regions. The MXD1 involvement in rRNA synthesis was also suggested by the nucleolar segregation upon rRNA synthesis inhibition by actinomycin D. Silencing of MXD1 with siRNAs resulted in increased synthesis of pre-rRNA while enforced MXD1 expression reduces it. The results suggest a new role for MXD1, which is the control of ribosome biogenesis. This new MXD1 function would be important to curb MYC activity in tumor cells. INTRODUCTION MXD1 acts as antagonist of MYC transcriptional activity [5–7]. Conversely to MYC, MXD1 is readily expressed MXD1 (also known as MAD1) is a transcription in resting and differentiated cells [6, 8, 9]. Moreover, factor belonging to the MXD family of proteins [1]. MXD1 inhibits cell proliferation and antagonize MYC The MXD family (composed of MXD1, MXI1/MXD2, transforming activity [10–12]. MXD3, MXD4 and MNT) is part of the MYC-MAX- The nucleolus is the nuclear factory for rRNA MXD network of transcription factors. These factors synthesis, posttranscriptional processing of pre-rRNA contain the b-HLH-LZ domain by which they bind to and assembly of preribosomal particles. It is organized E-boxes in the promoter of their target genes to regulate around clusters of tandem repeats of RNA genes (rDNA) their transcription [2–4].Whereas MYC-MAX activates that encode the rRNAs 28S, 18S and 5.8S. The nucleolar transcription, MXD1-MAX represses transcription structure, composed of fibrillar centers (FCs), dense forming complexes with histone deacetylases. Therefore, fibrillar component (DFC) and granular component www.impactjournals.com/oncotarget 69536 Oncotarget (GC), reflects the sequential steps of ribosome biogenesis of neurons was demonstrated by co-localization with [13–15]. The FC concentrates components of the UBF (Figure 1A, lower panel). Moreover, immunogold transcription machinery, such as RNA polymerase I and electron microscopy confirmed the localization of UBF (Upstream Binding Factor), an essential transcription MXD1 in FCs and associated DFC of the neuronal factor of ribosomal genes [16]. The DFC partially nucleolus (Figure 2B). To further confirm this subnuclear surrounds FCs and contains transcriptionally active distribution, we transfected HeLa cells with a GFP-MXD1 ribosomal genes and numerous RNA-binding proteins. construct. As controls, we also transfected a GFP-MNT RNA pol I transcription takes place in the FCs and in and GFP constructs. Following 24 h of transfection, the the DFC, with further posttranscriptional processing of transfected GFP-MXD1 protein was diffusely distributed pre-rRNA occurring within the DFC to give rise to the in the nucleoplasm, but concentrated in the nucleolus mature rRNAs [17, 18]. The GC is where the 40S and 60S (Figure 3A). The nucleolar localization of exogenous ribosome subunits are assembled [19, 20]. GFP-MXD1 was confirmed by immunofluorescence with MXD1 was described as nuclear in fibroblasts [1] the anti-UBF antibody, showing the co-localization of and quiescent keratinocytes [21], but the subcellular GFP and UBF signals (Figure 3B). localization of endogenous MXD1 is unknown for most We next studied whether the nucleolar distribution cell types. Here we report that MXD1 localizes in the of MXD1 was extensible to MYC and another member cell nucleolus and describe an interaction of MXD1 with of the MXD family as MNT. In sharp contrast to MXD1, UBF. In addition, we show that MXD1 binds to rDNA and both MYC and MNT were present in the nucleoplasm regulates rDNA transcription. but excluded from the UBF-immunolabeled nucleoli in most cells (Figure 4). Fluorescence intensity line profiles RESULTS confirmed the absence of MYC and MNT in the nucleolus (Supplementary Figure S2). Moreover, overexpressed MNT MXD1 localization in the nucleolus did not accumulate in the nucleolus, as shown in HeLa cells transfected with a GFP-MNT construct (Figure 3A). We performed immunofluorescence studies in The co-localization of MXD1 and UBF in the FCs proliferating embryonic stem cells HS181, mesenchymal opened the possibility that MXD1 might be regulating stem cells MSC-3H, myeloid leukemia K562 cells, rDNA transcription. To test this, we treated HS181 and embryonic kidney HEK293T cells, and cervical cancer MSC-3H cells with actinomycin D at low concentration HeLa cells. Although it is reported that MXD1 is mostly (50 ng/ml for 1 h), and performed co-immunostaining for expressed in quiescent cells or differentiated cells, MXD1 and UBF. Under these conditions, actinomycin we found low but detectable MXD1 protein levels in D inhibits RNA pol I but not RNA pol II and RNA proliferating cells of the above cell lines (Figure 1A), pol III [24, 25]. We also treated rats systemically although serum deprivation resulted in increased MXD1 with actinomycin (300 µg/kg for 3 h) and analysed expression, as described [22] (Figure 1A). We chose the nucleolar reorganization in sensory neurons from some of these cell lines with higher MXD1 expression to trigeminal ganglia. Interestingly, we found that RNA study its subcellular localization. As shown in Figure 1B, pol I inhibition interfered with the nucleolar targeting MXD1 was localized in the nucleus in all cell lines but, of MXD1 in all cases, as a MXD1-free nucleolar area surprisingly, it was also concentrated in the nucleolus in appeared after treatment with actinomycin D, meanwhile a relevant proportion of cells (70% to 100% depending the nucleoplasmic signal of MXD1 remained unchanged on the cell line). The nucleolar distribution of MXD1 was (Figure 5). In MSC-3H cells and sensory neurons, MXD1 confirmed by its co-localization with UBF (Figure 1B), was segregated at the nucleolar periphery, forming typical a nucleolar marker of FCs [13, 23]. The nucleolar perinucleolar caps or rings in confocal images (Figure 5) localization for MXD1 is so far unreported and we set [26]. This effect was associated with disruption of FCs out to confirm this. Analysis of the fluorescence intensity and peripheral segregation of the UBF, suggesting that profiles of UBF and MXD1 in cell lines and neurons MXD1 localization in FCs correlates with ongoing rRNA demonstrated this co-localization (Supplementary synthesis and raising a possible role of this transcription Figure S1). We also asked if the subnuclear localization factor in the regulation of rDNA transcription. of MXD1 observed in cells in culture also occurred in primary tissue cells. We analysed by immunofluorescence Interaction between MXD1 and UBF Sertoli and germinal cells from rat seminiferous tube and rat sensory ganglion neurons. MXD1 was concentrated As UBF and MXD1 co-localized in the FCs of in the nucleolus, with a weaker nucleoplasmic signal, nucleolus, we asked whether MXD1 and UBF interacted in post-mitotic Sertoli cells and neurons (Figure 2A). in the nucleolus. We first performed immunoprecipitation The nucleolar localization of MXD1 was confirmed assays in HeLa cells. The cells were deprived from serum in preparations co-stained with propidium iodide for 48 h to augment MXD1 expression and the lysates (Figure 2A), and its preferential concentration in FCs were immunoprecipitated with anti-UBF antibodies. www.impactjournals.com/oncotarget 69537 Oncotarget Figure 1: Endogenous MXD1 localizes in the nucleolus. (A) MXD1 protein expression in growing cells from different cell lines used in this work. MXD1 levels
Recommended publications
  • The Rise and Fall of the Bovine Corpus Luteum
    University of Nebraska Medical Center DigitalCommons@UNMC Theses & Dissertations Graduate Studies Spring 5-6-2017 The Rise and Fall of the Bovine Corpus Luteum Heather Talbott University of Nebraska Medical Center Follow this and additional works at: https://digitalcommons.unmc.edu/etd Part of the Biochemistry Commons, Molecular Biology Commons, and the Obstetrics and Gynecology Commons Recommended Citation Talbott, Heather, "The Rise and Fall of the Bovine Corpus Luteum" (2017). Theses & Dissertations. 207. https://digitalcommons.unmc.edu/etd/207 This Dissertation is brought to you for free and open access by the Graduate Studies at DigitalCommons@UNMC. It has been accepted for inclusion in Theses & Dissertations by an authorized administrator of DigitalCommons@UNMC. For more information, please contact [email protected]. THE RISE AND FALL OF THE BOVINE CORPUS LUTEUM by Heather Talbott A DISSERTATION Presented to the Faculty of the University of Nebraska Graduate College in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy Biochemistry and Molecular Biology Graduate Program Under the Supervision of Professor John S. Davis University of Nebraska Medical Center Omaha, Nebraska May, 2017 Supervisory Committee: Carol A. Casey, Ph.D. Andrea S. Cupp, Ph.D. Parmender P. Mehta, Ph.D. Justin L. Mott, Ph.D. i ACKNOWLEDGEMENTS This dissertation was supported by the Agriculture and Food Research Initiative from the USDA National Institute of Food and Agriculture (NIFA) Pre-doctoral award; University of Nebraska Medical Center Graduate Student Assistantship; University of Nebraska Medical Center Exceptional Incoming Graduate Student Award; the VA Nebraska-Western Iowa Health Care System Department of Veterans Affairs; and The Olson Center for Women’s Health, Department of Obstetrics and Gynecology, Nebraska Medical Center.
    [Show full text]
  • Uncovering the Signaling Landscape Controlling Breast Cancer Cell Migration Identifies Novel Metastasis Driver Genes
    ARTICLE https://doi.org/10.1038/s41467-019-11020-3 OPEN Uncovering the signaling landscape controlling breast cancer cell migration identifies novel metastasis driver genes Esmee Koedoot1,4, Michiel Fokkelman 1,4, Vasiliki-Maria Rogkoti1,4, Marcel Smid2, Iris van de Sandt1, Hans de Bont1, Chantal Pont1, Janna E. Klip1, Steven Wink 1, Mieke A. Timmermans2, Erik A.C. Wiemer2, Peter Stoilov3, John A. Foekens2, Sylvia E. Le Dévédec 1, John W.M. Martens 2 & Bob van de Water1 1234567890():,; Ttriple-negative breast cancer (TNBC) is an aggressive and highly metastatic breast cancer subtype. Enhanced TNBC cell motility is a prerequisite of TNBC cell dissemination. Here, we apply an imaging-based RNAi phenotypic cell migration screen using two highly motile TNBC cell lines (Hs578T and MDA-MB-231) to provide a repository of signaling determinants that functionally drive TNBC cell motility. We have screened ~4,200 target genes individually and discovered 133 and 113 migratory modulators of Hs578T and MDA-MB-231, respectively, which are linked to signaling networks predictive for breast cancer progression. The splicing factors PRPF4B and BUD31 and the transcription factor BPTF are essential for cancer cell migration, amplified in human primary breast tumors and associated with metastasis-free survival. Depletion of PRPF4B, BUD31 and BPTF causes primarily down regulation of genes involved in focal adhesion and ECM-interaction pathways. PRPF4B is essential for TNBC metastasis formation in vivo, making PRPF4B a candidate for further drug development. 1 Division of Drug Discovery and Safety, LACDR, Leiden University, Einsteinweg 55, Leiden 2333 CC, Netherlands. 2 Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam 3008 AE, Netherlands.
    [Show full text]
  • Activated Peripheral-Blood-Derived Mononuclear Cells
    Transcription factor expression in lipopolysaccharide- activated peripheral-blood-derived mononuclear cells Jared C. Roach*†, Kelly D. Smith*‡, Katie L. Strobe*, Stephanie M. Nissen*, Christian D. Haudenschild§, Daixing Zhou§, Thomas J. Vasicek¶, G. A. Heldʈ, Gustavo A. Stolovitzkyʈ, Leroy E. Hood*†, and Alan Aderem* *Institute for Systems Biology, 1441 North 34th Street, Seattle, WA 98103; ‡Department of Pathology, University of Washington, Seattle, WA 98195; §Illumina, 25861 Industrial Boulevard, Hayward, CA 94545; ¶Medtronic, 710 Medtronic Parkway, Minneapolis, MN 55432; and ʈIBM Computational Biology Center, P.O. Box 218, Yorktown Heights, NY 10598 Contributed by Leroy E. Hood, August 21, 2007 (sent for review January 7, 2007) Transcription factors play a key role in integrating and modulating system. In this model system, we activated peripheral-blood-derived biological information. In this study, we comprehensively measured mononuclear cells, which can be loosely termed ‘‘macrophages,’’ the changing abundances of mRNAs over a time course of activation with lipopolysaccharide (LPS). We focused on the precise mea- of human peripheral-blood-derived mononuclear cells (‘‘macro- surement of mRNA concentrations. There is currently no high- phages’’) with lipopolysaccharide. Global and dynamic analysis of throughput technology that can precisely and sensitively measure all transcription factors in response to a physiological stimulus has yet to mRNAs in a system, although such technologies are likely to be be achieved in a human system, and our efforts significantly available in the near future. To demonstrate the potential utility of advanced this goal. We used multiple global high-throughput tech- such technologies, and to motivate their development and encour- nologies for measuring mRNA levels, including massively parallel age their use, we produced data from a combination of two distinct signature sequencing and GeneChip microarrays.
    [Show full text]
  • Vitamin D Receptor As a Master Regulator of the C-MYC/MXD1 Network
    Vitamin D receptor as a master regulator of the c-MYC/MXD1 network Reyhaneh Salehi-Tabara, Loan Nguyen-Yamamotoa, Luz E. Tavera-Mendozaa, Thomas Quailb, Vassil Dimitrovb, Beum-Soo Anb,c, Leon Glassb, David Goltzmana,b, and John H. Whitea,b,1 Departments of aMedicine and bPhysiology, McGill University, Montreal, QC, Canada H3G 1Y6; and cDepartment of Biomaterial Science, College of Natural Resources and Life Science, Pusan National University, Republic of Korea Edited* by Hector F. DeLuca, University of Wisconsin-Madison, Madison, WI, and approved October 2, 2012 (received for review June 12, 2012) Vitamin D signaling regulates cell proliferation and differentiation, a better understanding of potential molecular mechanisms of the and epidemiological data suggest that it functions as a cancer protective effects of vitamin D. chemopreventive agent, although the underlying mechanisms are The VDR is bound by hormonal 1,25-dihydroxyvitamin D poorly understood. Vitamin D signaling can suppress expression of (1,25D), which is produced from vitamin D by largely hepatic 25- genes regulated by c-MYC, a transcription factor that controls hydroxylation, followed by 1α-hydroxylation by widely expressed epidermal differentiation and cell proliferation and whose activity CYP27B1 (17, 18). Potential cancer preventive actions of 1,25D is frequently elevated in cancer. We show through cell- and signaling through the VDR can be explained in part by the direct animal-based studies and mathematical modeling that hormonal interaction of the VDR with FoxO transcription factors, leading 1,25-dihydroxyvitamin D (1,25D) and the vitamin D receptor (VDR) to 1,25D-stimulated FoxO DNA-binding and target gene regu- profoundly alter, through multiple mechanisms, the balance in lation (19).
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • MOCHI Enables Discovery of Heterogeneous Interactome Modules in 3D Nucleome
    Downloaded from genome.cshlp.org on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press MOCHI enables discovery of heterogeneous interactome modules in 3D nucleome Dechao Tian1,# , Ruochi Zhang1,# , Yang Zhang1, Xiaopeng Zhu1, and Jian Ma1,* 1Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA #These two authors contributed equally *Correspondence: [email protected] Contact To whom correspondence should be addressed: Jian Ma School of Computer Science Carnegie Mellon University 7705 Gates-Hillman Complex 5000 Forbes Avenue Pittsburgh, PA 15213 Phone: +1 (412) 268-2776 Email: [email protected] 1 Downloaded from genome.cshlp.org on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press Abstract The composition of the cell nucleus is highly heterogeneous, with different constituents forming complex interactomes. However, the global patterns of these interwoven heterogeneous interactomes remain poorly understood. Here we focus on two different interactomes, chromatin interaction network and gene regulatory network, as a proof-of-principle, to identify heterogeneous interactome modules (HIMs), each of which represents a cluster of gene loci that are in spatial contact more frequently than expected and that are regulated by the same group of transcription factors. HIM integrates transcription factor binding and 3D genome structure to reflect “transcriptional niche” in the nucleus. We develop a new algorithm MOCHI to facilitate the discovery of HIMs based on network motif clustering in heterogeneous interactomes. By applying MOCHI to five different cell types, we found that HIMs have strong spatial preference within the nucleus and exhibit distinct functional properties. Through integrative analysis, this work demonstrates the utility of MOCHI to identify HIMs, which may provide new perspectives on the interplay between transcriptional regulation and 3D genome organization.
    [Show full text]
  • 1 a Genome-Wide RNA Interference Screen Identifies New Regulators Of
    Downloaded from genome.cshlp.org on September 30, 2021 - Published by Cold Spring Harbor Laboratory Press A genome-wide RNA interference screen identifies new regulators of androgen receptor function in prostate cancer cells Keren Imberg-Kazdan1 Susan Ha1,2 Alex Greenfield3 Christopher S. Poultney3^ Richard Bonneau3 Susan K. Logan1,2,4 Michael J. Garabedian2, 3,4 Departments of Biochemistry and Molecular Pharmacology1, Urology2, Microbiology4, and NYU Cancer Institute5, New York University School of Medicine, 550 First Avenue, New York, NY 10016; 3Center for Genomics and Systems Biology, New York University, New York, New York 10003 ^ present address: Seaver Autism Center for Research and Treatment, Department of Psychiatry, Mount Sinai Medical Center, New York, New York 10029 Corresponding Author: Michael J. Garabedian Department of Microbiology NYU School of Medicine 550 First Avenue, New York, NY 10016 Phone 212 263-7662 Fax 212 263-8276 Email: [email protected] Running Title: Genome-wide RNAi screen for AR regulators Keywords: Androgen receptor, RNAi screen, transcriptional regulation, HIPK2, MED19, Mediator complex, prostate cancer 1 Downloaded from genome.cshlp.org on September 30, 2021 - Published by Cold Spring Harbor Laboratory Press Abstract The androgen receptor (AR) is a mediator of both androgen-dependent and castration- resistant prostate cancers. Identification of cellular factors affecting AR transcriptional activity could in principle yield new targets that reduce AR activity and combat prostate cancer, yet a comprehensive analysis of the genes required for AR-dependent transcriptional activity has not been determined. Using an unbiased genetic approach that takes advantage of the evolutionary conservation of AR signaling, we have conducted a genome-wide RNAi screen in Drosophila cells for genes required for AR transcriptional activity and applied the results to human prostate cancer cells.
    [Show full text]
  • Cardiac SARS‐Cov‐2 Infection Is Associated with Distinct Tran‐ Scriptomic Changes Within the Heart
    Cardiac SARS‐CoV‐2 infection is associated with distinct tran‐ scriptomic changes within the heart Diana Lindner, PhD*1,2, Hanna Bräuninger, MS*1,2, Bastian Stoffers, MS1,2, Antonia Fitzek, MD3, Kira Meißner3, Ganna Aleshcheva, PhD4, Michaela Schweizer, PhD5, Jessica Weimann, MS1, Björn Rotter, PhD9, Svenja Warnke, BSc1, Carolin Edler, MD3, Fabian Braun, MD8, Kevin Roedl, MD10, Katharina Scher‐ schel, PhD1,12,13, Felicitas Escher, MD4,6,7, Stefan Kluge, MD10, Tobias B. Huber, MD8, Benjamin Ondruschka, MD3, Heinz‐Peter‐Schultheiss, MD4, Paulus Kirchhof, MD1,2,11, Stefan Blankenberg, MD1,2, Klaus Püschel, MD3, Dirk Westermann, MD1,2 1 Department of Cardiology, University Heart and Vascular Center Hamburg, Germany. 2 DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck. 3 Institute of Legal Medicine, University Medical Center Hamburg‐Eppendorf, Germany. 4 Institute for Cardiac Diagnostics and Therapy, Berlin, Germany. 5 Department of Electron Microscopy, Center for Molecular Neurobiology, University Medical Center Hamburg‐Eppendorf, Germany. 6 Department of Cardiology, Charité‐Universitaetsmedizin, Berlin, Germany. 7 DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany. 8 III. Department of Medicine, University Medical Center Hamburg‐Eppendorf, Germany. 9 GenXPro GmbH, Frankfurter Innovationszentrum, Biotechnologie (FIZ), Frankfurt am Main, Germany. 10 Department of Intensive Care Medicine, University Medical Center Hamburg‐Eppendorf, Germany. 11 Institute of Cardiovascular Sciences,
    [Show full text]
  • Pflugers Final
    CORE Metadata, citation and similar papers at core.ac.uk Provided by Serveur académique lausannois A comprehensive analysis of gene expression profiles in distal parts of the mouse renal tubule. Sylvain Pradervand2, Annie Mercier Zuber1, Gabriel Centeno1, Olivier Bonny1,3,4 and Dmitri Firsov1,4 1 - Department of Pharmacology and Toxicology, University of Lausanne, 1005 Lausanne, Switzerland 2 - DNA Array Facility, University of Lausanne, 1015 Lausanne, Switzerland 3 - Service of Nephrology, Lausanne University Hospital, 1005 Lausanne, Switzerland 4 – these two authors have equally contributed to the study to whom correspondence should be addressed: Dmitri FIRSOV Department of Pharmacology and Toxicology, University of Lausanne, 27 rue du Bugnon, 1005 Lausanne, Switzerland Phone: ++ 41-216925406 Fax: ++ 41-216925355 e-mail: [email protected] and Olivier BONNY Department of Pharmacology and Toxicology, University of Lausanne, 27 rue du Bugnon, 1005 Lausanne, Switzerland Phone: ++ 41-216925417 Fax: ++ 41-216925355 e-mail: [email protected] 1 Abstract The distal parts of the renal tubule play a critical role in maintaining homeostasis of extracellular fluids. In this review, we present an in-depth analysis of microarray-based gene expression profiles available for microdissected mouse distal nephron segments, i.e., the distal convoluted tubule (DCT) and the connecting tubule (CNT), and for the cortical portion of the collecting duct (CCD) (Zuber et al., 2009). Classification of expressed transcripts in 14 major functional gene categories demonstrated that all principal proteins involved in maintaining of salt and water balance are represented by highly abundant transcripts. However, a significant number of transcripts belonging, for instance, to categories of G protein-coupled receptors (GPCR) or serine-threonine kinases exhibit high expression levels but remain unassigned to a specific renal function.
    [Show full text]
  • Regulation and Function of Ptf1a in the Developing Nervous System
    REGULATION AND FUNCTION OF PTF1A IN THE DEVELOPING NERVOUS SYSTEM APPROVED BY SUPERVISORY COMMITTEE Jane E. Johnson, Ph.D. Raymond J. MacDonald, Ph.D. Ondine Cleaver, Ph.D. Steven L. McKnight, Ph.D. DEDICATION Work of this nature never occurs in a vacuum and would be utterly impossible without the contributions of a great many. I would like to thank my fellow lab members and collaborators, both past and present, for all of their help over the past several years, my thesis committee for their guidance and encouragement, and of course Jane for giving me every opportunity to succeed. To my friends, family, and my dear wife Maria, your love and support have made all of this possible. REGULATION AND FUNCTION OF PTF1A IN THE DEVELOPING NERVOUS SYSTEM by DAVID MILES MEREDITH DISSERTATION Presented to the Faculty of the Graduate School of Biomedical Sciences The University of Texas Southwestern Medical Center at Dallas In Partial Fulfillment of the Requirements For the Degree of DOCTOR OF PHILOSOPHY The University of Texas Southwestern Medical Center at Dallas Dallas, Texas May, 2012 REGULATION AND FUNCTION OF PTF1A IN THE DEVELOPING NERVOUS SYSTEM DAVID MILES MEREDITH The University of Texas Southwestern Medical Center at Dallas, 2012 JANE E. JOHNSON, Ph.D. Basic helix-loop-helix transcription factors serve many roles in development, including regulation of neurogenesis. Many of these factors are activated in naïve neural progenitors and function to promote neuronal differentiation and cell-type specification. Ptf1a is a basic helix-loop-helix protein that is required for proper inhibitory neuron formation in several regions of the developing nervous system, including the spinal cord, cerebellum, retina, and hypothalamus.
    [Show full text]
  • Theranostics Targeting Mir-21 with NL101 Blocks C-Myc/Mxd1 Loop And
    Theranostics 2021, Vol. 11, Issue 7 3439 Ivyspring International Publisher Theranostics 2021; 11(7): 3439-3451. doi: 10.7150/thno.53561 Research Paper Targeting miR-21 with NL101 blocks c-Myc/Mxd1 loop and inhibits the growth of B cell lymphoma Shu Li1,2, Xin He1,3, Yichao Gan1,4, Jiawei Zhang1,2, Feiqiong Gao1, Limin Lin1,2, Xi Qiu1, Teng Yu1, Xuzhao Zhang1, Panpan Chen1, Jiefeng Tong1, Wenbin Qian1,5, Yang Xu1,5,6 1. Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China. 2. Zhejiang University Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China. 3. Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010. 4. Institute of Genetics, Zhejiang University and Department of Genetics, Zhejiang University School of Medicine, Hangzhou 310058, China. 5. National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 215006, China. 6. Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China. Corresponding author: Yang Xu, Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China. Tel: 86-571-89713679; Fax: 86-571-87783655; E-mail: [email protected]. © The author(s). This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/).
    [Show full text]
  • Datasheet A06485-1 Anti-MXD1 Antibody
    Product datasheet Anti-MXD1 Antibody Catalog Number: A06485-1 BOSTER BIOLOGICAL TECHNOLOGY Special NO.1, International Enterprise Center, 2nd Guanshan Road, Wuhan, China Web: www.boster.com.cn Phone: +86 27 67845390 Fax: +86 27 67845390 Email: [email protected] Basic Information Product Name Anti-MXD1 Antibody Gene Name MXD1 Source Rabbit IgG Species Reactivity human,mouse,rat Tested Application WB,Direct ELISA Contents 500ug/ml antibody with PBS ,0.02% NaN3 , 1mg BSA and 50% glycerol. Immunogen E. coli-derived human Mad recombinant protein (Position: M1-L221). Purification Immunogen affinity purified. Observed MW 28KD Dilution Ratios Western blot: 1:500-2000 Direct ELISA: 1:100-1000 Storage 12 months from date of receipt,-20℃ as supplied.6 months 2 to 8℃ after reconstitution. Avoid repeated freezing and thawing Background Information MAD protein is a protein that in humans is encoded by the MXD1 gene. This gene encodes a member of the MYC/MAX/MAD network of basic helix-loop-helix leucine zipper transcription factors. The MYC/MAX/MAD transcription factors mediate cellular proliferation, differentiation and apoptosis. The encoded protein antagonizes MYC-mediated transcriptional activation of target genes by competing for the binding partner MAX and recruiting repressor complexes containing histone deacetylases. Mutations in this gene may play a role in acute leukemia, and the encoded protein is a potential tumor suppressor. Alternatively spliced transcript variants encoding multiple isoforms have been observed for this gene. Reference Anti-MXD1 Antibody被引用在0文献中。 暂无引用 FOR RESEARCH USE ONLY. NOT FOR DIAGNOSTIC AND CLINICAL USE. 1 Product datasheet Anti-MXD1 Antibody Catalog Number: A06485-1 BOSTER BIOLOGICAL TECHNOLOGY Special NO.1, International Enterprise Center, 2nd Guanshan Road, Wuhan, China Web: www.boster.com.cn Phone: +86 27 67845390 Fax: +86 27 67845390 Email: [email protected] Selected Validation Data Figure 1.
    [Show full text]