Tbx3 Is a Downstream Target of the Wnt/B-Catenin Pathway and a Critical Mediator of B-Catenin Survival Functions in Liver Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Tbx3 Is a Downstream Target of the Wnt/B-Catenin Pathway and a Critical Mediator of B-Catenin Survival Functions in Liver Cancer Research Article Tbx3 Is a Downstream Target of the Wnt/B-Catenin Pathway and a Critical Mediator of B-Catenin Survival Functions in Liver Cancer Claire-Ange´lique Renard,1,2 Charlotte Labalette,1,2 Carolina Armengol,1,2 Delphine Cougot,1,2 Yu Wei,1,2 Stefano Cairo,1,2 Pascal Pineau,1,3 Christine Neuveut,1,2 Aure´lien de Reynie`s,4 Anne Dejean,1,3 Christine Perret,5,6 and Marie-Annick Buendia1,2 1Institut National de la Sante et de la Recherche Medicale, U579; 2Oncogenesis and Molecular Virology Unit and 3Nuclear Organization and Oncogenesis Unit, Institut Pasteur; 4Ligue Nationale contre le Cancer; 5De´partementGe´ne´tique,De´veloppementet Pathologie Mole´culaire, Institut Cochin; and 6Institut National de la Sante et de la Recherche Medicale, U567, Paris, France Abstract interacts with DNA-binding factors of the T-cell factor/lymphoid enhancer factor (Tcf/Lef) family to promote transcription of Wnt Tbx3 encodes a transcriptional repressor that is important for diverse patterning events during development, and Tbx3 target genes. Depending on cell context, the selective expression of mutation in humans causes the ulnar-mammary syndrome. distinct sets of Wnt-responsive genes is strictly controlled by the interplay of signaling pathways. Increasing numbers of candidate Here, we describe the identification of Tbx3 in array-based h search for genes downstream Wnt/B-catenin that are impli- -catenin targets include cell cycle and growth regulators and proteins involved in cell-matrix interactions, migration and cated in liver tumorigenesis. Overexpression of Tbx3 is closely 7 B invasion, and in the regulation of the Wnt pathway. associated with the mutational status of -catenin in murine h liver tumors induced by Myc as well as in human hepatocel- Recent studies have outlined the importance of Wnt/ -catenin lular carcinomas and hepatoblastomas. Moreover, Tbx3 signaling in regulating liver cell proliferation during development transcription is activated by ectopic expression of B-catenin (2–4) and in governing essential functions in the adult liver (5–7). in mouse liver and in human tumor cell lines. Evidence that Moreover, aberrant reactivation of Wnt signaling is a predominant B mechanism implicated in liver tumorigenesis. Mutations of the Tbx3 transcription is directly regulated by -catenin is h provided by chromatin immunoprecipitation and reporter -catenin and Axin genes leading to constitutive activation of h-catenin have been identified in hepatocellular carcinoma (HCC) assays. Although HepG2 cells stably transfected with Tbx3 display moderately enhanced growth rate, the dominant and hepatoblastoma, and frequent overexpression of the Wnt negative mutant Tbx3-Y149S drastically inhibits hepatoma receptor Frizzled-7 is a major early event in hepatocarcinogenesis in vitro in vivo (refs. 8–11; for reviews, see refs. 12, 13). Animal models have been cell growth and . Moreover, small interfering h RNAs (siRNA) directed against Tbx3 inhibit anchorage- instrumental in allowing the identification of -catenin target independent growth of liver and colon carcinoma cells. We genes in nondiseased or tumorous liver, including liver-specific further show that inhibition of Tbx3 expression by specific enzymes involved in glutamine and nitrogen metabolism, such as siRNAs blocks B-catenin–mediated cell survival and renders glutamine synthetase (GS), ornithine aminotransferase, the gluta- cells sensitive to doxorubicin-induced apoptosis. Conversely, mate transporter (GLT-1), and the cytochrome P450 enzymes ectopic expression of Tbx3 inhibits apoptosis induced by CYP1A2 and CYP2E1 (5, 6, 14–17). However, c-myc has not been found thus far to be regulated by h-catenin in the liver context, and B-catenin depletion. Marked overexpression of Tbx3 in a subset of hepatoblastomas is associated with chemotherapy- the oncogenic program triggered by Wnt signaling in hepatocarci- nogenesis remains largely unknown. resistant phenotype and unfavorable patient outcome. These Myc results reveal an unsuspected role of Tbx3 as a mediator of We have shown previously that transgenic mice carrying a B-catenin activities on cell proliferation and survival and as oncogene controlled by woodchuck hepatitis virus (WHV) regu- latory sequences were highly predisposed to liver cancer (18, 19). an important player in liver tumorigenesis. [Cancer Res h 2007;67(3):901–10] Activating mutations of -catenin were found at significant rates in these HCCs, as for other Myc transgenic strains, suggesting that the survival functions of h-catenin might rescue tumor cells from Introduction Myc-induced apoptosis (8, 19, 20). Consistent with this notion, The Wnt/Wingless pathway plays a pivotal role in embryonic inactivation of p53 was found to represent an alternative oncogenic development and adult homeostasis by regulating cell fate, mechanism (9, 19). To explore downstream genetic programs proliferation, and differentiation, and it is aberrantly reactivated activated by Wnt/h-catenin signaling in liver cancer, we employed in various human cancers (1). Mutations in adenomatous polyposis microarray profiling and found that the T-box protein 3 (Tbx3) was coli, Axin, and h-catenin genes that mimic Wnt activation result in specifically activated in murine tumors carrying mutant h-catenin. the stabilization of h-catenin, which then moves to the nucleus and The closely related genes TBX2 and TBX3 are members of the T-box gene family that play an important role in patterning events during development. Tbx3 is notably implicated in heart, limb, Note: Supplementary data for this article are available at Cancer Research Online and posterior digit specification and in mammary gland develop- (http://cancerres.aacrjournals.org/). Requests for reprints: Marie-Annick Buendia, Oncogenesis and Molecular ment (21, 22). In humans, Tbx3 mutations are responsible for the Virology Unit, Institut National de la Sante et de la Recherche Medicale U579, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France. Phone: 33-145-688-866; Fax: 33-145-688-943; E-mail: [email protected]. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-06-2344 7 http://www.stanford.edu/~rnusse/pathways/targets.html. www.aacrjournals.org 901 Cancer Res 2007; 67: (3). February 1, 2007 Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2007 American Association for Cancer Research. Cancer Research ulnar-mammary syndrome (UMS), an autosomal dominant disor- Total RNA was extracted from each sample using the RNeasy Kit der characterized by upper limb deficiencies and apocrine/ (Qiagen) and treated with RNase-free DNase (Ambion). mammary gland hypoplasia (23). Tbx2/Tbx3 contain a conserved Oligonucleotide microarray and statistical analysis. Fifty-five human transcription-repression domain and can repress basal and HCCs, 24 hepatoblastomas, and 9 pools of nontumorous livers (5 from adults and 4 from children) were selected on the basis of high RNA quality as activated transcription (24). Identification of p14ARF as a direct determined with the Agilent 2100 bioanalyzer (Agilent Technologies, Palo target of Tbx2/Tbx3 has linked these transcriptional repressors Alto, CA). Raw feature data from Affymetrix HG-U133A GeneChip micro- to the control of cell cycle and senescence and to the antipro- arrays were obtained for these samples. All samples were processed using the liferative response delivered by the ARF-Mdm2-p53 pathway in one-cycle target labeling protocol.8 We analyzed the data using the tumorigenesis (25, 26). R system software (v2.2.1), GeneSpring GX 7.3, and BRB ArrayTools (v3.3.1). In this study, we showed that the expression of Tbx3, but not Tbx2, Raw feature data were normalized using the Robust Multi-array method was induced by mutant h-catenin in different systems, including (R package affy, v1.4.32; ref. 27), which yielded log2 intensity expression normal murine liver, murine and human liver tumors, and human summary values for each of the 22,283 probe sets. Each set (HCC and carcinoma cell lines. We cloned the TBX3 promoter and used a hepatoblastoma) was normalized independently. Additional statistical anal- combination of chromatin immunoprecipitation (ChIP) and report- yses were done with SPSS 11.0 package (SPSS, Inc., Chicago, IL). Comparison between groups was done using the Fisher’s exact test. Probability of er assays that led to identify Tbx3 as a direct transcriptional target of h overall survival was determined using the Kaplan-Meier analysis and the log- the -catenin/Tcf complex. We also showed that Tbx3 plays a crucial rank and Breslow tests. Follow-up was closed at the time of death or last visit. role in cell proliferation and survival. Inhibition of Tbx3 expression Plasmids. The reporter plasmid L-Tbx3-Luc carrying the TBX3 promoter by RNA interference dramatically reduced anchorage-independent was generated by PCR amplification of human genomic DNA sequences growth and abolished h-catenin protection over doxorubicin- (University of California–Santa CruzGenome Bioinformatics Site) extend- induced apoptosis in tumor cells. Finally, we propose that strong ing 2,226 bp upstream and 619 bp downstream of the transcriptional start overexpression of Tbx3 contributes to the chemotherapy-resistant site of the human TBX3 gene (GenBank accession number NM_016569). We phenotype of a subset of hepatoblastomas. used the following primers: L-Tbx3F, 5¶-CTAGCTAGCGAAACCCTGCAGT- GACTTCCG-3¶; and L-Tbx3R, 5¶-GGAAGATCTGCTCGAAATAGACACTC- CAGC-3¶. The PCR product was cloned into pGL3-Basic
Recommended publications
  • 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]
  • Tbx3 Controls the Sinoatrial Node Gene Program and Imposes Pacemaker Function on the Atria
    Downloaded from genesdev.cshlp.org on September 29, 2021 - Published by Cold Spring Harbor Laboratory Press Tbx3 controls the sinoatrial node gene program and imposes pacemaker function on the atria Willem M.H. Hoogaars,1,3 Angela Engel,2,3 Janynke F. Brons,1,3 Arie O. Verkerk,2 Frederik J. de Lange,1 L.Y. Elaine Wong,1 Martijn L. Bakker,1 Danielle E. Clout,1 Vincent Wakker,1 Phil Barnett,1 Jan Hindrik Ravesloot,2 Antoon F.M. Moorman,1 E. Etienne Verheijck,2 and Vincent M. Christoffels1,4 1Department of Anatomy and Embryology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; 2Department of Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands The sinoatrial node initiates the heartbeat and controls the rate and rhythm of contraction, thus serving as the pacemaker of the heart. Despite the crucial role of the sinoatrial node in heart function, the mechanisms that underlie its specification and formation are not known. Tbx3, a transcriptional repressor required for development of vertebrates, is expressed in the developing conduction system. Here we show that Tbx3 expression delineates the sinoatrial node region, which runs a gene expression program that is distinct from that of the bordering atrial cells. We found lineage segregation of Tbx3-negative atrial and Tbx3-positive sinoatrial node precursor cells as soon as cardiac cells turn on the atrial gene expression program. Tbx3 deficiency resulted in expansion of expression of the atrial gene program into the sinoatrial node domain, and partial loss of sinoatrial node-specific gene expression.
    [Show full text]
  • TBX3 Acts As Tissue-Specific Component of the Wnt/Β
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.22.053561; this version posted April 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. TBX3 acts as tissue-specific component of the Wnt/b-catenin enhanceosome Dario Zimmerli1,6#, Costanza Borrelli2#, Amaia Jauregi-Miguel3,4#, Simon Söderholm3,4, Salome Brütsch1, Nikolaos Doumpas1, Jan Reichmuth1, Fabienne Murphy-Seiler5, Michel Aguet5, Konrad Basler1*, Andreas E. Moor2*, Claudio Cantù3,4* 1 Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland, CH-8057 2 Institute of Molecular Cancer Research, University of Zurich, Zürich, Switzerland, CH-8057 3 Wallenberg Centre for Molecular Medicine, Linköping University 4 Department of Biomedical and Clinical Sciences, Faculty of Health Science, SE-581 83 Linköping, Sweden 5Swiss Institute for Experimental Cancer Research (ISREC), Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, CH-1015 Lausanne, Switzerland 6 Current address: Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands # These authors contributed equally to this work * For correspondence: [email protected] [email protected] [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.22.053561; this version posted April 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license.
    [Show full text]
  • The Role of Tbx5 in Sinoatrial Node Differentiation in Mouse Embryonic Stem Cell Derived Cardiomyocytes Yunkai Dai Clemson University, [email protected]
    Clemson University TigerPrints All Dissertations Dissertations May 2019 The Role of Tbx5 in Sinoatrial Node Differentiation in Mouse Embryonic Stem Cell Derived Cardiomyocytes Yunkai Dai Clemson University, [email protected] Follow this and additional works at: https://tigerprints.clemson.edu/all_dissertations Recommended Citation Dai, Yunkai, "The Role of Tbx5 in Sinoatrial Node Differentiation in Mouse Embryonic Stem Cell Derived Cardiomyocytes" (2019). All Dissertations. 2377. https://tigerprints.clemson.edu/all_dissertations/2377 This Dissertation is brought to you for free and open access by the Dissertations at TigerPrints. It has been accepted for inclusion in All Dissertations by an authorized administrator of TigerPrints. For more information, please contact [email protected]. THE ROLE OF TBX5 IN SINOATRIAL NODE DIFFERENTIATION IN MOUSE EMBRYONIC STEM CELL DERIVED CARDIOMYOCYTES A Dissertation Presented to the Graduate School of Clemson University In Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy Bioengineering by Yunkai Dai May 2019 Accepted by: Dr Ann Foley, Committee Chair Dr Agneta Simionescu Dr Robin C. Muise-Helmericks Dr Ying Mei ABSTRACT The sinoatrial node of the mouse embryo arises from the wall of the right atrium near the border of the sinus venosus. Early in development this region expresses the transcription factor Tbx5. Because of this, Tbx5 is thought to sit at the apex of a transcriptional cascade leading to sinoatrial node (SAN) differentiation. To test this we produced a mouse embryonic stem cell line B1 (pTripZ-mTbx5; αMHC::GFP) that conditionally overexpresses Tbx5, to determine if this would lead to enhanced SAN differentiation. We found that ES cells overexpressing Tbx5 showed enhanced overall cardiac differentiation and that cardiac cells showed increased beat rates as compared control embryos.
    [Show full text]
  • Differential Spatio-Temporal Regulation of T-Box Gene Expression by Micrornas During Cardiac Development
    Journal of Cardiovascular Development and Disease Article Differential Spatio-Temporal Regulation of T-Box Gene Expression by microRNAs during Cardiac Development Mohamad Alzein, Estefanía Lozano-Velasco , Francisco Hernández-Torres , Carlos García-Padilla , Jorge N. Domínguez , Amelia Aránega and Diego Franco * Cardiovascular Development Group, Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; [email protected] (M.A.); [email protected] (E.L.-V.); [email protected] (F.H.-T.); [email protected] (C.G.-P.); [email protected] (J.N.D.); [email protected] (A.A.) * Correspondence: [email protected] Abstract: Cardiovascular development is a complex process that starts with the formation of symmet- rically located precardiac mesodermal precursors soon after gastrulation and is completed with the formation of a four-chambered heart with distinct inlet and outlet connections. Multiple transcrip- tional inputs are required to provide adequate regional identity to the forming atrial and ventricular chambers as well as their flanking regions; i.e., inflow tract, atrioventricular canal, and outflow tract. In this context, regional chamber identity is widely governed by regional activation of distinct T-box family members. Over the last decade, novel layers of gene regulatory mechanisms have been discovered with the identification of non-coding RNAs. microRNAs represent the most well-studied subcategory among short non-coding RNAs. In this study, we sought to investigate the functional role of distinct microRNAs that are predicted to target T-box family members. Our data demonstrated a highly dynamic expression of distinct microRNAs and T-box family members during cardiogenesis, revealing a relatively large subset of complementary and similar microRNA–mRNA expression pro- Citation: Alzein, M.; Lozano-Velasco, files.
    [Show full text]
  • The Combined Action of Esrrb and Nr5a2 Is Essential for Naïve Pluripotency
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.05.134999; this version posted June 6, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. The combined action of Esrrb and Nr5a2 is essential for naïve pluripotency. Nicola Festuccia1,2,*, Nick Owens3, Almira Chervova2, Agnès Dubois2, and Pablo Navarro2 1MRC London Institute of Medical Sciences (LMS), Imperial College London, Du Cane Road, W12 0NN London, UK 2Epigenomics, Proliferation, and the Identity of Cells, Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR3738, 75015 Paris, France 3Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter EX2 5DW, UK *Corresponding author: [email protected] The maintenance of pluripotency in mouse embryonic stem cells both during early differentiation (Festuccia et al., 2018a) (ESCs) is governed by the action of an interconnected network and, conversely, during reprogramming to induced pluripo- of transcription factors. Among them, only Oct4 and Sox2 have tency (Adachi et al., 2018); it orchestrates the recruitment of been shown to be strictly required for the self-renewal of ESCs the transcriptional machinery (Bell et al., 2020; Percharde and pluripotency, particularly in culture conditions where dif- et al., 2012) and other transcription factors (Adachi et al., ferentiation cues are chemically inhibited. Here, we report that 2018) to key regulatory elements of the pluripotency network the conjunct activity of two orphan nuclear receptors, Esrrb (Whyte et al., 2013); it maintains its binding activity dur- and Nr5a2, parallels the importance of that of Oct4 and Sox2 in naïve ESCs.
    [Show full text]
  • Systematic Review and Meta-Analysis of Candidate Gene Association Studies of Lower Urinary Tract Symptoms in Men
    S D E U Eur Urol. 2014 Oct; 66(4): 752–768. PMCID: PMC4410299 doi: 10.1016/j.eururo.2014.01.007 Systematic Review and Meta-analysis of Candidate Gene Association Studies of Lower Urinary Tract Symptoms in Men ☆ Rufus Cartwright,a,†⁎ Altaf Mangera,b,† Kari A.O. Tikkinen,c Prabhakar Rajan,d Jori Pesonen,e Anna C. Kirby,f Ganesh Thiagamoorthy,g Chris Ambrose,h Juan Gonzalez-Maffe,i Phillip R. Bennett,j Tom Palmer,k Andrew Walley,l Marjo-Riitta Järvelin,m Vik Khullar,n and Chris Chappleo a Department of Epidemiology and Biostatistics and Department of Urogynaecology, Imperial College London, London, UK b Department of Urology Research, University of Sheffield, Sheffield, UK c Department of Urology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland, and Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada d Beatson Institute for Cancer Research, University of Glasgow, Glasgow, UK e Department of Urology, Tampere University Hospital, and School of Medicine, University of Tampere, Tampere, Finland f Department of Reproductive Medicine, University of California, San Diego, San Diego, CA, USA, and Department of Obstetrics and Gynecology, Kaiser Permanente, San Diego, CA, USA g Department of Urogynaecology, King's College London, London, UK h University College London Medical School, London, UK i Clinical Trials Unit, Imperial College London, London, UK j Institute for Reproductive and Developmental Biology, Imperial College London, London, UK k Division of Health Sciences,
    [Show full text]
  • Life-Threatening Cardiac Episode in a Polish Patient Carrying Contiguous Gene Microdeletion of the TBX5 and the TBX3 Genes
    Iwanicka‑Pronicka et al. SpringerPlus (2016) 5:1638 DOI 10.1186/s40064-016-3275-1 CASE STUDY Open Access Life‑threatening cardiac episode in a Polish patient carrying contiguous gene microdeletion of the TBX5 and the TBX3 genes Katarzyna Iwanicka‑Pronicka1,2*, Magdalena Socha3, Maria Jędrzejowska1, Małgorzata Krajewska‑Walasek1 and Aleksander Jamsheer3,4 Abstract Holt–Oram syndrome (HOS) features radial ray hypoplasia, heart defect and cardiac conduction impairment. Ulnar- mammary syndrome (UMS) characterizes congenital defects of the ulnar side of the upper limbs, underdevelop‑ ment of apocrine glands including hypoplasia and the dysfunction of mammary glands, hypogonadism and obesity. Inheritance of both conditions is autosomal dominant, mutations or deletions are found in the TBX5 and TBX3 gene, respectively. The Polish patient presented short stature, obesity, congenital malformation of the radial and ulnar side of the upper limbs, heart block, hypogonadism and dysmorphic features. At the age of 13 years he lost conscious‑ ness developing respiratory insufficiency caused by bradycardia in the course of sudden atrioventricular third degree heart block requiring immediate implantation of pace maker-defibrillator device. Microdeletion of the 12q24.21 was identified using array CGH method. This region includes contiguous genes the TBX5, TBX3, and part of RBM19. The patient initially diagnosed as having HOS, was found to present the UMS features as well. Array CGH method should be applied in patients suspected of HOS or UMS, especially
    [Show full text]
  • Breast Ductal Carcinoma in Situ Carry Mutational Driver Events
    Modern Pathology (2017) 30, 952–963 OPEN 952 Official journal of the United States and Canadian Academy of Pathology Breast ductal carcinoma in situ carry mutational driver events representative of invasive breast cancer Jia-Min B Pang1,2, Peter Savas3, Andrew P Fellowes1, Gisela Mir Arnau4, Tanjina Kader5,6, Ravikiran Vedururu1, Chelsee Hewitt1, Elena A Takano1, David J Byrne1, David YH Choong1, Ewan KA Millar7,8,9,10, C Soon Lee10,11,12, Sandra A O’Toole7,11,13, Sunil R Lakhani14,15, Margaret C Cummings14,15, G Bruce Mann16,17, Ian G Campbell5,6, Alexander Dobrovic18, Sherene Loi3,5, Kylie L Gorringe2,5,6,19 and Stephen B Fox1,2,5,19 1Department of Pathology, Peter MacCallum Cancer Centre, Grattan Street, Melbourne, VIC, Australia; 2Department of Pathology, University of Melbourne, Grattan Street, Parkville, Melbourne, VIC, Australia; 3Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, Grattan Street, Melbourne, VIC, Australia; 4Molecular Genomics Core, Peter MacCallum Cancer Centre, Grattan Street, Melbourne, VIC, Australia; 5Sir Peter MacCallum Department of Oncology, University of Melbourne, Grattan Street, Parkville, Melbourne, VIC, Australia; 6Cancer Genomics Program, Peter MacCallum Cancer Centre, Grattan Street, Melbourne, VIC, Australia; 7Translational Breast Cancer Research, The Kinghorn Cancer Centre and Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; 8Department of Anatomical Pathology, South Eastern Area Pathology Service, St George Hospital, Kogarah, NSW, Australia; 9School of Medical
    [Show full text]
  • Estrogen Expands Breast Cancer Stem-Like Cells Through Paracrine FGF/Tbx3 Signaling
    Estrogen expands breast cancer stem-like cells through paracrine FGF/Tbx3 signaling Christine M. Fillmorea,b, Piyush B. Guptac,1, Jenny A. Rudnickb,d, Silvia Caballerob,d, Patricia J. Kellerb,d, Eric S. Landerc,e, and Charlotte Kuperwassera,b,d,2 aGenetics Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111; bMolecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111; cBroad Institute of MIT/Harvard and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142; dDepartment of Anatomy and Cellular Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111; and eDepartment of Systems Biology, Harvard Medical School, Boston, MA 02115 Edited by Geoffrey M. Wahl, Salk Institute for Biological Studies, La Jolla, CA, and accepted by the Editorial Board October 26, 2010 (received for review June 17, 2010) Many tumors contain heterogeneous populations of cells, only cells but, rather, activates their proliferation through paracrine some of which exhibit increased tumorigenicity and resistance to signaling (14, 15). These data imply a two-component mammary + anticancer therapies. Evidence suggests that these aggressive stem cell niche in which estrogen signaling in the ERα non- cancer cells, often termed “cancer stem cells” or “cancer stem-like stem cell compartment stimulates the proliferation of cells within α− cells” (CSCs), rely upon developmental signaling pathways that are the ER stem cell compartment. In breast cancer, it is unclear whether stem-like cells are also important for survival and expansion of normal stem cells. Here fi we report that, in analogy to embryonic mammary epithelial bi- regulated by speci c hormone-growth factor paracrine signal- ology, estrogen signaling expands the pool of functional breast ing pathways.
    [Show full text]
  • Estrogen Receptor B As a Therapeutic Target in Breast Cancer Stem Cells
    JNCI J Natl Cancer Inst (2017) 109(3): djw236 doi: 10.1093/jnci/djw236 First published online February 10, 2017 Article ARTICLE Estrogen Receptor b as a Therapeutic Target in Breast Cancer Stem Cells Ran Ma, Govindasamy-Muralidharan Karthik, John Lo¨ vrot, Felix Haglund, Gustaf Rosin, Anne Katchy, Xiaonan Zhang, Lisa Viberg, Jan Frisell, Cecilia Williams, Stig Linder, Irma Fredriksson, Johan Hartman Affiliations of authors: Department of Oncology and Pathology (RM, GMK, JL, FH, XZ, LV, SL, JH), Cancer Center Karolinska (RM, GMK, JL, FH, GR, XZ, LV, SL, JH), and Department of Molecular Medicine and Surgery (JF, IF), Karolinska Institutet, Stockholm, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (GR, CW); Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX (AK, CW); Department of Pathology and Cytology, Karolinska University Laboratory, Stockholm, Sweden (FH, JH); Department of Breast and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden (JF, IF); Science for Life Laboratory, Department of Proteomics, KTH, Royal Institute of Technology, Stockholm, Sweden (CW); Department of Medical and Health Sciences, Department of Medicine and Health, Linko¨ ping University, Linko¨ ping, Sweden (SL); Correspondence to: Johan Hartman, M.D., PhD, Department of Oncology and Pathology, Karolinska Institutet, 17176 Stockholm, Sweden (e-mail: [email protected]). Abstract Abstract Background: Breast cancer cells with tumor-initiating capabilities (BSCs) are considered to maintain tumor growth and govern metastasis. Hence, targeting BSCs will be crucial to achieve successful treatment of breast cancer. Methods: We characterized mammospheres derived from more than 40 cancer patients and two breast cancer cell lines for the expression of estrogen receptors (ERs) and stem cell markers.
    [Show full text]
  • Metabolic Network-Based Stratification of Hepatocellular Carcinoma Reveals Three Distinct Tumor Subtypes
    Metabolic network-based stratification of hepatocellular carcinoma reveals three distinct tumor subtypes Gholamreza Bidkhoria,b,1, Rui Benfeitasa,1, Martina Klevstigc,d, Cheng Zhanga, Jens Nielsene, Mathias Uhlena, Jan Borenc,d, and Adil Mardinoglua,b,e,2 aScience for Life Laboratory, KTH Royal Institute of Technology, SE-17121 Stockholm, Sweden; bCentre for Host-Microbiome Interactions, Dental Institute, King’s College London, SE1 9RT London, United Kingdom; cDepartment of Molecular and Clinical Medicine, University of Gothenburg, SE-41345 Gothenburg, Sweden; dThe Wallenberg Laboratory, Sahlgrenska University Hospital, SE-41345 Gothenburg, Sweden; and eDepartment of Biology and Biological Engineering, Chalmers University of Technology, SE-41296 Gothenburg, Sweden Edited by Sang Yup Lee, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea, and approved November 1, 2018 (received for review April 27, 2018) Hepatocellular carcinoma (HCC) is one of the most frequent forms of of markers associated with recurrence and poor prognosis (13–15). liver cancer, and effective treatment methods are limited due to Moreover, genome-scale metabolic models (GEMs), collections tumor heterogeneity. There is a great need for comprehensive of biochemical reactions, and associated enzymes and transporters approaches to stratify HCC patients, gain biological insights into have been successfully used to characterize the metabolism of subtypes, and ultimately identify effective therapeutic targets. We HCC, as well as identify drug targets for HCC patients (11, 16–18). stratified HCC patients and characterized each subtype using tran- For instance, HCC tumors have been stratified based on the uti- scriptomics data, genome-scale metabolic networks and network lization of acetate (11). Analysis of HCC metabolism has also led topology/controllability analysis.
    [Show full text]