Deregulation of the HOXA10 Homeobox Gene in Endometrial Carcinoma: Role in Epithelial-Mesenchymal Transition

Total Page:16

File Type:pdf, Size:1020Kb

Deregulation of the HOXA10 Homeobox Gene in Endometrial Carcinoma: Role in Epithelial-Mesenchymal Transition Research Article Deregulation of the HOXA10 Homeobox Gene in Endometrial Carcinoma: Role in Epithelial-Mesenchymal Transition Hiroyuki Yoshida,1 Russell Broaddus,2 Wenjun Cheng,1 SuSu Xie,2 and Honami Naora1 Departments of 1Molecular Therapeutics and 2Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas Abstract normally expressed during early urogenital development, and is expressed in renal tumors (6). PAX2 is thought to promote cell Homeobox genes encode transcription factors that control cell survival, and inhibiting PAX2 expression in cancer cells induces differentiation and play essential roles in developmental apoptosis (7). On the other hand, many homeobox genes that are patterning. Increasing evidence indicates that many homeo- normally expressed in differentiated tissues are lost or down- box genes are aberrantly expressed in cancers, and that their regulated in tumors. Nkx3.1 controls prostate morphogenesis and deregulation significantly contributes to tumor progression. is normally expressed in the prostate during development and in The homeobox gene HOXA10 controls uterine organogenesis the adult (8). NKX3.1 is frequently deleted in prostate cancers (9), during embryonic development and functional endometrial and enforced expression of NKX3.1 in prostate cancer cells differentiation in the adult. We investigated whether HOXA10 inhibits cell growth (10). Many other homeobox genes contribute expression is deregulated in endometrial carcinomas, and to tumor progression by promoting cell growth and/or survival how counteracting this aberrant expression modifies tumor (4, 5). However, given the important role of homeobox genes in behavior. We found that down-regulation of HOXA10 expres- cell differentiation, surprisingly little is known regarding their role sion in endometrial carcinomas strongly correlates with in EMT. increased tumor grade and is associated with methylation of Endometrial carcinoma is the fourth most frequently diagnosed the HOXA10 promoter. Enforced expression of HOXA10 in cancer in women. Grade 1 (G ) endometrioid carcinomas of the endometrial carcinoma cells inhibited invasive behavior 1 endometrium are typically well-differentiated, and are mostly in vitro and tumor dissemination in nude mice. The inhibitory detected while still confined to the uterine corpus (11). Grade 3 effect of HOXA10 on invasive behavior was attributable at (G ) endometrioid carcinomas are mostly solid, whereas uterine least in part to the ability of HOXA10 to induce expression of 3 papillary serous carcinoma (UPSC) is characterized by papillary the epithelial cell adhesion molecule E-cadherin by down- patterns of growth. Although histologically distinct, G endome- regulating expression of Snail, a repressor of E-cadherin gene 3 trioid carcinoma and UPSC share several common features transcription. These findings reveal a novel role for HOXA10 including high-grade nuclear atypia and poor responsiveness to deregulation in the progression of endometrial carcinoma by chemotherapy, radiation, and progestin therapy (11–13). G promoting epithelial-mesenchymal transition. (Cancer Res 3 endometrioid carcinoma and UPSC have greater propensity for 2006; 66(2): 889-97) myometrial and lymphatic/vascular space invasion than G1 tumors (11–13), but the mechanisms underlying the progression of these Introduction high-grade tumors are poorly understood. It is widely recognized that many of the molecular pathways Several studies indicate that the HOXA10 homeobox gene that underlie tumor progression are aberrations of processes that controls uterine organogenesis during embryonic development control normal embryonic development. Epithelial-mesenchymal and functional endometrial differentiation in the adult. HOXA10 transition (EMT) is a dynamic process whereby epithelial cells expression becomes restricted to the uterine primordium during lose polarity and cell-cell adhesion, acquire a migratory pheno- normal mu¨llerian duct differentiation, and is maintained in the type, and switch to a mesenchymal-like gene expression program. adult uterus (14). Female mice with a targeted disruption of This process is essential for normal morphogenesis, and reactiva- Hoxa10 produce normal embryos, but their uteri are unable to tion of the EMT program is regarded as a primary mechanism support embryo implantation (15, 16). Implantation normally underlying carcinoma progression and invasive behavior (1, 2). occurs during the mid-secretory phase of the menstrual cycle, a Homeobox genes have increasingly been thought to also represent period during which the endometrium undergoes extensive important hubs in the intimate relationship between tumor glandular differentiation and HOXA10 expression in the endome- progression and embryogenesis. These genes encode transcription trial glands dramatically increases (14). A role for HOXA10 in factors that control developmental patterning and specify the endometrial differentiation is also supported by our recent findings unique morphologic identities of adult tissues (3, 4). Several that aberrant activation of HOXA10 in ovarian surface epithelium- homeobox genes that are normally expressed in embryonic tissues derived cells gives rise to the endometrial-like features of are activated in tumors (4, 5). One example is PAX2, which is endometrioid ovarian carcinoma (17). Because HOXA10 plays an essential role in endometrial development, we speculated that its expression is deregulated in endometrial carcinomas. In this study, we found that down- regulation of HOXA10 expression in endometrial carcinomas Requests for reprints: Honami Naora, Department of Molecular Therapeutics, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, correlates with increased tumor grade and is associated with Box 184, Houston, TX 77030. Phone: 713-563-4222; Fax: 713-563-4235; E-mail: hnaora@ HOXA10 promoter methylation. Enforced expression of HOXA10 in mdanderson.org. I2006 American Association for Cancer Research. endometrial carcinoma cells inhibited invasive behavior, and this doi:10.1158/0008-5472.CAN-05-2828 inhibitory effect was attributable, at least in part, to the ability of www.aacrjournals.org 889 Cancer Res 2006; 66: (2). January 15, 2006 Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2006 American Association for Cancer Research. Cancer Research HOXA10 to up-regulate E-cadherin by suppressing the expression Absorbance at 570 nm after MTT addition was measured daily over 5-day of the Snail transcription factor. These findings reveal that time course. The number of trypan blue–excluding cells in replicate plates silencing of HOXA10 expression in the endometrium could were also directly counted by using a hemocytometer. Each assay was done contribute to tumor progression by promoting EMT. in triplicate. Reverse transcription-PCR. Reverse transcription using 1 Ag of DNase I-treated total RNA was done using Superscript II reverse transcriptase Materials and Methods (Invitrogen) in a reaction volume of 20 AL. One microliter of the reverse Cell lines, cDNAs, and antibodies. The endometrial carcinoma cell transcription reaction was used for amplification using Platinum Taq DNA lines SPEC2 and KLE were obtained from Isaiah Fidler (University of polymerase (Invitrogen). Detection of HOXA10 and actin expression has Texas M.D. Anderson Cancer Center, Houston, TX) and American Type been previously described (17). Other primers are as follows: (Snail) sense, Culture Collection (Rockville, MD), respectively. The HOXA10 cDNA clone 5V-TGCGCGAATCGGCGACCC-3V; antisense, 5V-CCTAGAGAAGGCCTTCC- was provided by Corey Largman (University of California VA Medical CGCAG-3V; (Slug) sense, 5V-GCCTCCAAAAAGCCAAACTACAG-3V; antisense, Center, San Francisco, CA). The rabbit polyclonal antibody to HOXA10 has 5V-GTGTGCTACACAGCAGCC-3V. Amplification was done as follows: dena- been previously described (17). Other antibodies used are as follows: actin, turation at 94jC for 1 minute, annealing at 61jC for 1 minute and extension h1 integrin, cadherin-6, Snail (Santa Cruz Biotechnology, Santa Cruz, CA), at 72jC for 2 minutes for 37 cycles to detect Snail expression, and E-cadherin (Zymed Laboratories, San Francisco, CA), vimentin (Neo- denaturation at 95jC for 1 minute, annealing at 58jC for 1 minute and Markers, Fremont, CA), Ki-67 (Vector Laboratories, Burlingame, CA). extension at 72jC for 1 minute for 40 cycles to detect Slug expression. Secondary antibodies were obtained from Kirkegaard & Perry Laboratories Titrations were done to ensure a linear range of amplification. (Gaithersburg, MD). Western blot and immunofluorescence staining. Cell lysates were Tissue microarrays and immunohistochemistry. Tissue microarrays prepared by using M-PER buffer (Pierce, Rockford, IL). Equal amounts of were constructed from core biopsies of endometrial carcinoma specimens protein (20 Ag per lane) were separated by SDS-PAGE and transferred to at the University of Texas M.D. Anderson Cancer Center using a Beecher polyvinylidene difluoride membranes (Amersham Pharmacia Biotech, Instruments (Silver Spring, MD) tissue arrayer. Use of specimens was Piscataway, NJ). Membranes were probed with antibodies to E-cadherin approved by the Institutional Review Board. Cases included the following: (1 Ag/mL), h1 integrin (1:100), cadherin-6 (1:200), and actin (1:100). For cell normal secretory endometrium (n = 10), G1 (n = 26), G2 (n = 37), G3 (n = 58) staining, cells were fixed in 4% paraformaldehyde and permeabilized
Recommended publications
  • Detection of Interacting Transcription Factors in Human Tissues Using
    Myšičková and Vingron BMC Genomics 2012, 13(Suppl 1):S2 http://www.biomedcentral.com/1471-2164/13/S1/S2 PROCEEDINGS Open Access Detection of interacting transcription factors in human tissues using predicted DNA binding affinity Alena Myšičková*, Martin Vingron From The Tenth Asia Pacific Bioinformatics Conference (APBC 2012) Melbourne, Australia. 17-19 January 2012 Abstract Background: Tissue-specific gene expression is generally regulated by combinatorial interactions among transcription factors (TFs) which bind to the DNA. Despite this known fact, previous discoveries of the mechanism that controls gene expression usually consider only a single TF. Results: We provide a prediction of interacting TFs in 22 human tissues based on their DNA-binding affinity in promoter regions. We analyze all possible pairs of 130 vertebrate TFs from the JASPAR database. First, all human promoter regions are scanned for single TF-DNA binding affinities with TRAP and for each TF a ranked list of all promoters ordered by the binding affinity is created. We then study the similarity of the ranked lists and detect candidates for TF-TF interaction by applying a partial independence test for multiway contingency tables. Our candidates are validated by both known protein-protein interactions (PPIs) and known gene regulation mechanisms in the selected tissue. We find that the known PPIs are significantly enriched in the groups of our predicted TF-TF interactions (2 and 7 times more common than expected by chance). In addition, the predicted interacting TFs for studied tissues (liver, muscle, hematopoietic stem cell) are supported in literature to be active regulators or to be expressed in the corresponding tissue.
    [Show full text]
  • Prospective Isolation of NKX2-1–Expressing Human Lung Progenitors Derived from Pluripotent Stem Cells
    The Journal of Clinical Investigation RESEARCH ARTICLE Prospective isolation of NKX2-1–expressing human lung progenitors derived from pluripotent stem cells Finn Hawkins,1,2 Philipp Kramer,3 Anjali Jacob,1,2 Ian Driver,4 Dylan C. Thomas,1 Katherine B. McCauley,1,2 Nicholas Skvir,1 Ana M. Crane,3 Anita A. Kurmann,1,5 Anthony N. Hollenberg,5 Sinead Nguyen,1 Brandon G. Wong,6 Ahmad S. Khalil,6,7 Sarah X.L. Huang,3,8 Susan Guttentag,9 Jason R. Rock,4 John M. Shannon,10 Brian R. Davis,3 and Darrell N. Kotton1,2 2 1Center for Regenerative Medicine, and The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA. 3Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA. 4Department of Anatomy, UCSF, San Francisco, California, USA. 5Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA. 6Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, Massachusetts, USA. 7Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA. 8Columbia Center for Translational Immunology & Columbia Center for Human Development, Columbia University Medical Center, New York, New York, USA. 9Department of Pediatrics, Monroe Carell Jr. Children’s Hospital, Vanderbilt University, Nashville, Tennessee, USA. 10Division of Pulmonary Biology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA. It has been postulated that during human fetal development, all cells of the lung epithelium derive from embryonic, endodermal, NK2 homeobox 1–expressing (NKX2-1+) precursor cells.
    [Show full text]
  • Reciprocal Regulation of TEAD4 and CCN2 for the Trophectoderm Development of the Bovine Blastocyst
    155 6 REPRODUCTIONRESEARCH Reciprocal regulation of TEAD4 and CCN2 for the trophectoderm development of the bovine blastocyst Hiroki Akizawa1,2, Ken Kobayashi3, Hanako Bai1, Masashi Takahashi1, Shinjiro Kagawa1, Hiroaki Nagatomo1,† and Manabu Kawahara1 1Laboratory of Animal Genetics and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan, 2Japan Society for the Promotion of Science (JSPS Research Fellow), Tokyo, Japan and 3Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan Correspondence should be addressed to M Kawahara; Email: [email protected] †(Hiroaki Nagatomo is now at Department of Biotechnology, Faculty of Life and Environmental Science, University of Yamanashi, Kofu, Japan) Abstract The first segregation at the blastocyst stage is the symmetry-breaking event to characterize two cell components; namely, inner cell mass (ICM) and trophectoderm (TE). TEA domain transcription factor 4 (TEAD4) is a well-known regulator to determine TE properties of blastomeres in rodent models. However, the roles of bovine TEAD4 in blastocyst development have been unclear. We here aimed to clarify the mechanisms underlining TE characterization by TEAD4 in bovine blastocysts. We first found that the TEAD4 mRNA expression level was greater in TE than in ICM, which was further supported by TEAD4 immunofluorescent staining. Subsequently, we examined the expression patterns of TE-expressed genes; CDX2, GATA2 and CCN2, in the TEAD4-knockdown (KD) blastocysts. These expression levels significantly decreased in the TEAD4 KD blastocysts compared with controls. Of these downregulated genes, the CCN2 expression level decreased the most. We further analyzed the expression levels of TE-expressed genes; CDX2, GATA2 and TEAD4 in the CCN2 KD blastocysts.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Regulation of the Tumor Suppressor Homeogene Cdx2 by Hnf4a in Intestinal Cancer
    Oncogene (2013) 32, 3782–3788 & 2013 Macmillan Publishers Limited All rights reserved 0950-9232/13 www.nature.com/onc SHORT COMMUNICATION Regulation of the tumor suppressor homeogene Cdx2 by HNF4a in intestinal cancer T Saandi1,2, F Baraille3,4,5, L Derbal-Wolfrom1,2, A-L Cattin3,4,5, F Benahmed1,6, E Martin1,2, P Cardot3,4,5, B Duclos1,2,7, A Ribeiro3,4,5, J-N Freund1,2,8 and I Duluc1,2,8 The gut-specific homeotic transcription factor Cdx2 is a crucial regulator of intestinal development and homeostasis, which is downregulated in colorectal cancers (CRC) and exhibits a tumor suppressor function in the colon. We have previously established that several endodermal transcription factors, including HNF4a and GATA6, are involved in Cdx2 regulation in the normal gut. Here we have studied the role of HNF4a in the mechanism of deregulation of Cdx2 in colon cancers. Crossing ApcD14/ þ mice prone to spontaneous intestinal tumor development with pCdx2-9LacZ transgenic mice containing the LacZ reporter under the control of the 9.3-kb Cdx2 promoter showed that this promoter segment contains sequences recapitulating the decrease of Cdx2 expression in intestinal cancers. Immunohistochemistry revealed that HNF4a, unlike GATA6, exhibited a similar decrease to Cdx2 in genetic (Apcmin/ þ and ApcD14/ þ ) and chemically induced (Azoxymethane (AOM) treatment) models of intestinal tumors in mice. HNF4a and Cdx2 also exhibited a comparable deregulated pattern in human CRC. Correlated patterns were observed between HNF4a and Cdx2 in several experimental models of human colon cancer cell lines: xenografts in nude mice, wound healing and glucose starvation.
    [Show full text]
  • Role of Cdx Factors in Early Mesodermal Fate Decisions Tanya E
    © 2019. Published by The Company of Biologists Ltd | Development (2019) 146, dev170498. doi:10.1242/dev.170498 RESEARCH ARTICLE Role of Cdx factors in early mesodermal fate decisions Tanya E. Foley, Bradley Hess, Joanne G. A. Savory, Randy Ringuette and David Lohnes* ABSTRACT Cdx1−/− or Cdx2+/− phenotypes, indicative of functional overlap Murine cardiac and hematopoietic progenitors are derived from Mesp1+ between Cdx family members (Faas and Isaacs, 2009; Savory et al., mesoderm. Cdx function impacts both yolk sac hematopoiesis and 2011; van den Akker et al., 2002; van Nes et al., 2006; Young et al., Cdx2 cardiogenesis in zebrafish, suggesting that Cdx family members 2009). The derivation and analysis of conditional mutants regulate early mesoderm cell fate decisions. We found that Cdx2 (Savory et al., 2009a; Stringer et al., 2012) and compound mutant occupies a number of transcription factor loci during embryogenesis, derivatives (van den Akker et al., 2002; Savory et al., 2011; Verzi including key regulators of both cardiac and blood development, and et al., 2011; van Rooijen et al., 2012; Stringer et al., 2012; Grainger that Cdx function is required for normal expression of the cardiogenic et al., 2010; Hryniuk et al., 2012) further substantiated the transcription factors Nkx2-5 and Tbx5. Furthermore, Cdx and Brg1, an functional overlap between family members. Analysis of these ATPase subunit of the SWI/SNF chromatin remodeling complex, co- mutants also revealed a requirement for Cdx genes in diverse occupy a number of loci, suggesting that Cdx family members regulate processes in development and in the adult intestinal track. These target gene expression through alterations in chromatin architecture.
    [Show full text]
  • Association of Gene Ontology Categories with Decay Rate for Hepg2 Experiments These Tables Show Details for All Gene Ontology Categories
    Supplementary Table 1: Association of Gene Ontology Categories with Decay Rate for HepG2 Experiments These tables show details for all Gene Ontology categories. Inferences for manual classification scheme shown at the bottom. Those categories used in Figure 1A are highlighted in bold. Standard Deviations are shown in parentheses. P-values less than 1E-20 are indicated with a "0". Rate r (hour^-1) Half-life < 2hr. Decay % GO Number Category Name Probe Sets Group Non-Group Distribution p-value In-Group Non-Group Representation p-value GO:0006350 transcription 1523 0.221 (0.009) 0.127 (0.002) FASTER 0 13.1 (0.4) 4.5 (0.1) OVER 0 GO:0006351 transcription, DNA-dependent 1498 0.220 (0.009) 0.127 (0.002) FASTER 0 13.0 (0.4) 4.5 (0.1) OVER 0 GO:0006355 regulation of transcription, DNA-dependent 1163 0.230 (0.011) 0.128 (0.002) FASTER 5.00E-21 14.2 (0.5) 4.6 (0.1) OVER 0 GO:0006366 transcription from Pol II promoter 845 0.225 (0.012) 0.130 (0.002) FASTER 1.88E-14 13.0 (0.5) 4.8 (0.1) OVER 0 GO:0006139 nucleobase, nucleoside, nucleotide and nucleic acid metabolism3004 0.173 (0.006) 0.127 (0.002) FASTER 1.28E-12 8.4 (0.2) 4.5 (0.1) OVER 0 GO:0006357 regulation of transcription from Pol II promoter 487 0.231 (0.016) 0.132 (0.002) FASTER 6.05E-10 13.5 (0.6) 4.9 (0.1) OVER 0 GO:0008283 cell proliferation 625 0.189 (0.014) 0.132 (0.002) FASTER 1.95E-05 10.1 (0.6) 5.0 (0.1) OVER 1.50E-20 GO:0006513 monoubiquitination 36 0.305 (0.049) 0.134 (0.002) FASTER 2.69E-04 25.4 (4.4) 5.1 (0.1) OVER 2.04E-06 GO:0007050 cell cycle arrest 57 0.311 (0.054) 0.133 (0.002)
    [Show full text]
  • CDX2 As a Useful Marker of Colorectal Adenocarcinoma Metastases to Lung in Pre-Operative Biopsy Specimens
    87-92 1/6/07 12:56 Page 87 ONCOLOGY REPORTS 18: 87-92, 2007 87 CDX2 as a useful marker of colorectal adenocarcinoma metastases to lung in pre-operative biopsy specimens SHIGEBUMI TANAKA1, KANA SAITO1,2, TOMOKAZU ITO1, KOHEI TAJIMA3, AKIRA MOGI3, YOSHINORI SHITARA4, TAKAAKI SANO2 and HIROYUKI KUWANO1 Departments of 1General Surgical Science, 2Tumor Pathology, Gunma Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511; 3Department of Surgery, Fujioka General Hospital, 942-1 Fujioka, Fujioka, Gunma 375-8503; 4Department of Surgery, Isesaki General Hospital, 12-1 Tsunatorihoncho, Isesaki, Gunma 372-0821, Japan Received January 12, 2007; Accepted April 17, 2007 Abstract. Although distinguishing metastatic colorectal as a solitary nodule. Differential diagnosis between metastatic adenocarcinoma from primary lung adenocarcinoma is often adenocarcinoma of colorectal origin and primary lung adeno- difficult, pre- or intra-operative identification is very important, carcinoma using imaging techniques such as chest computed as the resection areas for each diagnosis differ substantially. tomography (CT) is thus often difficult in patients with a CDX2, a recently cloned homeobox gene, represents a highly history of colorectal cancer. However, this differentiation is specific and sensitive marker of colorectal adenocarcinoma. crucial for therapeutic and prognostic purposes (1). Particularly We evaluated CDX2 expression using pre- and intra-operative in cases with operative indications, pre- or intra-operative biopsy specimens. The study examined 50 consecutive colo- identification is very important, as the resection areas differ rectal adenocarcinoma metastases to the lung, including 20 substantially between the diagnoses. If the tumor is a solitary biopsy specimens and 66 resected specimens, and 21 primary metastatic colorectal carcinoma to the lung, partial lung lung adenocarcinomas.
    [Show full text]
  • 1714 Gene Comprehensive Cancer Panel Enriched for Clinically Actionable Genes with Additional Biologically Relevant Genes 400-500X Average Coverage on Tumor
    xO GENE PANEL 1714 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes 400-500x average coverage on tumor Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11 IL6R KAT5 ACVR2A
    [Show full text]
  • Immunohistochemistry Stain Offerings
    immunohistochemistry stain offerings TRUSTED PATHOLOGISTS. INVALUABLE ANSWERS.™ MARCHMAY 20172021 www.aruplab.com/ap-ihcaruplab.com/ap-ihc InformationInformation in this brochurein this brochure is current is current as of as May of March 2021. 2017. All content All content is subject is subject to tochange. change. Please contactPlease ARUPcontact ClientARUP Services Client Services at 800-522-2787 at (800) 522-2787 with any with questions any questions or concerns.or concerns. ARUP LABORATORIES As a nonprofit, academic institution of the University of Utah and its Department We believe in of Pathology, ARUP believes in collaborating, sharing and contributing to laboratory science in ways that benefit our clients and their patients. collaborating, Our test menu is one of the broadest in the industry, encompassing more sharing and than 3,000 tests, including highly specialized and esoteric assays. We offer comprehensive testing in the areas of genetics, molecular oncology, pediatrics, contributing pain management, and more. to laboratory ARUP’s clients include many of the nation’s university teaching hospitals and children’s hospitals, as well as multihospital groups, major commercial science in ways laboratories, and group purchasing organizations. We believe that healthcare should be delivered as close to the patient as possible, which is why we support that provide our clients’ efforts to be the principal healthcare provider in the communities they serve by offering highly complex assays and accompanying consultative support. the best value Offering analytics, consulting, and decision support services, ARUP provides for the patient. clients with the utilization management tools necessary to prosper in this time of value-based care.
    [Show full text]
  • HOXB7 Is an Erα Cofactor in the Activation of HER2 and Multiple ER Target Genes Leading
    Author Manuscript Published OnlineFirst on July 15, 2015; DOI: 10.1158/2159-8290.CD-15-0090 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. HOXB7 is an ERα cofactor in the activation of HER2 and multiple ER target genes leading to endocrine resistance Kideok Jin1, Sunju Park1, Wei Wen Teo1, Preethi Korangath1, Sean Soonweng Cho1, Takahiro Yoshida1*, Balázs Győrffy2, Chirayu Pankaj Goswami3#, Harikrishna Nakshatri3, Leigh-Ann Cruz1, Weiqiang Zhou4, Hongkai Ji4, Ying Su5, Muhammad Ekram5, Zhengsheng Wu6, Tao Zhu6, Kornelia Polyak5, and Saraswati Sukumar1≠ 1Breast Cancer Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 22nd Department of Pediatrics, MTA TTK Lendület Cancer Biomarker Research Group, Semmelweis University, Budapest, Hungary, 3Center for Computational Biology and Bioinformatics, Indiana University, IN, 4Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD and 5Dana- Farber Cancer Institute, Boston, MA. 6Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, People's Republic of China. Current address: *Department of Surgery, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan #Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA ≠ To whom correspondence may be addressed: Saraswati Sukumar, PhD Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans Street, CRB 1, Room 143, Baltimore, MD 21231-1000. Phone: 410-614-2479 1 Downloaded from cancerdiscovery.aacrjournals.org on September 28, 2021. © 2015 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 15, 2015; DOI: 10.1158/2159-8290.CD-15-0090 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
    [Show full text]