The Receptor Interacting Protein 1 Inhibits P53 Induction Through NF-KB Activation and Confers a Worse Prognosis in Glioblastoma

Total Page:16

File Type:pdf, Size:1020Kb

The Receptor Interacting Protein 1 Inhibits P53 Induction Through NF-KB Activation and Confers a Worse Prognosis in Glioblastoma Research Article The Receptor Interacting Protein 1 Inhibits p53 Induction through NF-KB Activation and Confers a Worse Prognosis in Glioblastoma Seongmi Park,1 Kimmo J. Hatanpaa,2,7 Yang Xie,3,8 Bruce E. Mickey,4,7 Christopher J. Madden,4,7 Jack M. Raisanen,2,7 Deepti B. Ramnarain,1 Guanghua Xiao,3 Debabrata Saha,5 David A. Boothman,8 Dawen Zhao,6 Robert M. Bachoo,1,7,8 Russell O. Pieper,9 and Amyn A. Habib1,7,8 Departments of 1Neurology, 2Pathology, 3Clinical Sciences, 4Neurosurgery, 5Radiation Oncology, and 6Radiology, 7Annette G. Strauss Center of Neurooncology, and 8Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas and 9Department of Neurological Surgery, University of California-San Francisco, San Francisco, California Abstract studies have linked components of the NF-nB signaling pathway to Nuclear factor-KB (NF-KB) activation may play an important cell cycle progression and tumorigenesis (11–16). role in the pathogenesis of cancer and also in resistance to An intriguing mechanism underlying the pathogenesis of treatment. Inactivation of the p53 tumor suppressor is a key inflammation-induced cancer is the negative regulation of tumor component of the multistep evolution of most cancers. Links suppressor pathways by inflammatory and stress-induced signals. between the NF-KB and p53 pathways are under intense p53 is a key tumor suppressor altered in a broad range of human investigation. In this study, we show that the receptor cancers, including glioma, and an important outcome of p53 interacting protein 1 (RIP1), a central component of the activation is cell cycle arrest or apoptosis after DNA damage K (17, 18). Previous studies have documented links between the NF- NF- B signaling network, negatively regulates p53 tumor sup- n pressor signaling. Loss of RIP1 from cells results in augmented B and p53 networks that have largely been reported to be induction of p53 in response to DNA damage, whereas antagonistic (19) but may also be synergistic (20). There is evidence that components of the NF-nB signaling network interact with p53 increased RIP1 level leads to a complete shutdown of DNA h damage–induced p53 induction by enhancing levels of cellular at multiple levels. For example, IKK2 (IKK ) inhibits p53 induction in response to chemotherapeutic drugs via an up-regulation of mdm2. The key signal generated by RIP1 to up-regulate mdm2 a and inhibit p53 is activation of NF-KB. The clinical implication mdm2 (21), whereas IKK1 (IKK ) interferes with p53-mediated gene transcription by inducing CBP phosphorylation (22). Thus, of this finding is shown in glioblastoma, the most common n primary malignant brain tumor in adults. We show that RIP1 NF- B pathway–mediated inhibition of p53 function may promote is commonly overexpressed in glioblastoma, but not in grades the pathogenesis of cancer. II and III glioma, and increased expression of RIP1 confers a The death domain–containing kinase receptor interacting worse prognosis in glioblastoma. Importantly, RIP1 levels protein 1 (RIP1, RIPK1) is an essential component of the signaling n correlate strongly with mdm2 levels in glioblastoma. Our cascade that activates NF- B in response to cellular stress and results show a key interaction between the NF-KB and p53 inflammation (23, 24). Thus, RIP1 is required for tumor necrosis a a n pathways that may have implications for the targeted factor (TNF )–induced and DNA damage–induced NF- B treatment of glioblastoma. [Cancer Res 2009;69(7):2809–16] activation (25–28). In addition, RIP1 is also a key component of innate immunity, essential for TLR-3–mediated activation of NF-nB (29). RIP1 is composed of kinase, intermediate, and death domains. Introduction RIP1 is involved in the activation of the IKKs via a kinase- A link between chronic inflammation and cancer has been independent mechanism (30). Thus, RIP1 seems to function as an suspected for over a century (1). A major link between adaptor, and it is the intermediate domain of RIP1 that is essential n inflammation and cancer is mediated by activation of nuclear for NF- B activation. factor-nB(NF-nB; refs. 2–4). NF-nB activation is a strong Glioblastoma (glioblastoma multiforme) is the most common prosurvival signal (5). Constitutive and deregulated activation of primary malignant brain tumor in adults and is resistant to NF-nB is widespread in human cancer (3, 6) and promotes survival treatment (31). The median survival of glioblastoma patients with of tumor cells and resistance to treatment (7, 8). Furthermore, radiation and chemotherapy was recently noted to be 14.6months experimental models support a causal role for NF-nB activation in (32). The molecular pathogenesis of glioblastoma includes genetic inflammation-induced cancer (9, 10). Cross-talk between stress- alterations in pathways mediating proliferation, apoptosis, and cell induced/inflammatory responses and oncogenic signaling path- cycle control (33, 34). Inflammatory responses can be readily ways is likely to play an important role in cancer. A number of detected in glioma in the form of infiltrating macrophages/ microglia and lymphocytes, production of inflammatory cytokines, and activation of NF-nB (35, 36). Importantly, NF-nB activation Note: Supplementary data for this article are available at Cancer Research Online may be linked to the resistance of glioblastoma cells to (http://cancerres.aacrjournals.org/). O6-alkylating agents (37, 38). S. Park and K.J. Hatanpaa contributed equally to this work. Requests for reprints: Amyn A. Habib, University of Texas Southwestern Medical In this study, we show that RIP1 negatively regulates p53 Center, Mail Code 8813, 6001 Forest Park ND4.136, Dallas, TX 75390-8813. Phone: 214- induction in response to DNA damage. RIP1 regulates p53 via the 645-6237; Fax: 214-645-6240; E-mail: [email protected]. I2009 American Association for Cancer Research. up-regulation of mdm2 levels. We also elucidate a key role for doi:10.1158/0008-5472.CAN-08-4079 NF-nB activation in RIP1-mediated regulation of mdm2 and p53 www.aacrjournals.org 2809 Cancer Res 2009; 69: (7). April 1, 2009 Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 2009 American Association for Cancer Research. Cancer Research down-regulation. Analysis of glioma patient samples showed that low groups based on a RIP1 value of 1.2, which represents a 50% increase RIP1 is overexpressed in f30% of glioblastoma (grade IV), but not in over the levels found in normal brain. To investigate whether RIP1 level grades II and III glioma. Importantly, there was a striking correlation (ratio between RIP1 and ERK2) is associated with the clinical outcome, we between RIP1 and mdm2 levels in glioblastoma, consistent with our applied a proportional hazard model using both overall survival time and progression-free survival time as the outcomes (censored by the last known mechanistic data in vitro. Finally, high RIP1 level is an independent number of follow-up months). The multivariate proportional hazard model negative prognostic indicator in glioblastoma. was used to analyze the effect of additional prognosis factors, such as age and Karnofsky scores, with the RIP1 level. The survival curves were fitted using the Kaplan-Meier method. The log-rank test was used to compare Materials and Methods different survival curves. Fisher’s exact test was used to calculate the P value Plasmids and cell lines. RIP1 plasmid was obtained from Dr. Brian Seed for association between RIP1 and mdm2 in glioblastoma. The survival and cloned into pcDNA3.1 with a COOH terminal FLAG tag using standard analysis was performed using the survival package in SAS software. +/+ À/À molecular techniques. rip1 and rip1 murine embryo fibroblasts were provided by Dr. Michelle Kelliher (27). In this study, we used primary mouse embryo fibroblasts (MEF) at passages 3 to 4. A p21-LUC promoter was Results provided by Dr. Bert Vogelstein. RIP1 influences p53 induction in response to DNA damage. Antibodies, reagents, and Western blotting. p53 (DO1), InBa, p65, and Previous studies have documented interactions between compo- extracellular signal-regulated kinase 2 (ERK2) antibodies were obtained n from Santa Cruz Biotechnology. The RIP antibody was purchased from BD nents of the NF- B signaling network and p53. Thus, we Biosciences and was used for both immunohistochemistry and Western investigated whether the cellular level of RIP1 could influence À/À blotting. mdm2 (2A10) antibodies were purchased from Calbiochem. p53 p53 induction in response to DNA damage. Exposure of RIP1 (1C12), p21, and FLAG were purchased from Cell Signaling Technology. We primary MEFs to ionizing radiation (IR) resulted in a significantly used p53 DO1 antibodies for the detection of p53 in (human) glioma cell greater induction of p53 compared with wild-type cells (Fig. 1A). À À lines, whereas p53 1C12 antibodies were used for the detection of (mouse) Similarly, exposure of RIP1 / MEFs to UV radiation elicited a p53 in MEFs. For tumor samples, 30 Ag of protein were loaded on to 7.5% greater induction of p53 (Supplementary Fig. S1A). Next, we polyacrylamide gels, and Western blotting was performed according to investigated the effect of increased RIP1 expression in the human standard protocols. glioblastoma cell line U87MG, which expresses wild-type p53. To Luciferase assays. U87MG cells (70K) were plated in 24-well dishes overexpress RIP1, we used a tetracycline-inducible adenoviral followed by transfection with either p21-LUC along with RIP plasmid or empty vector, using calcium phosphate. A dual-luciferase reporter assay expression vector (tet-off). An empty adenoviral vector (Ad-null) C system was used according to the instructions of the manufacturer was used as an additional control (Supplementary Fig. S1 ). Cells (Promega). Firefly luciferase activity was measured in a luminometer and were exposed to IR (10 Gy) and then followed by Western blot. normalized on the basis of Renilla luciferase activity.
Recommended publications
  • Homeobox Gene Expression Profile in Human Hematopoietic Multipotent
    Leukemia (2003) 17, 1157–1163 & 2003 Nature Publishing Group All rights reserved 0887-6924/03 $25.00 www.nature.com/leu Homeobox gene expression profile in human hematopoietic multipotent stem cells and T-cell progenitors: implications for human T-cell development T Taghon1, K Thys1, M De Smedt1, F Weerkamp2, FJT Staal2, J Plum1 and G Leclercq1 1Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospital, Ghent, Belgium; and 2Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands Class I homeobox (HOX) genes comprise a large family of implicated in this transformation proces.14 The HOX-C locus transcription factors that have been implicated in normal and has been primarily implicated in lymphomas.15 malignant hematopoiesis. However, data on their expression or function during T-cell development is limited. Using degener- Hematopoietic cells are derived from stem cells that reside in ated RT-PCR and Affymetrix microarray analysis, we analyzed fetal liver (FL) in the embryo and in the adult bone marrow the expression pattern of this gene family in human multipotent (ABM), which have the unique ability to self-renew and thereby stem cells from fetal liver (FL) and adult bone marrow (ABM), provide a life-long supply of blood cells. T lymphocytes are a and in T-cell progenitors from child thymus. We show that FL specific type of hematopoietic cells that play a major role in the and ABM stem cells are similar in terms of HOX gene immune system. They develop through a well-defined order of expression, but significant differences were observed between differentiation steps in the thymus.16 Several transcription these two cell types and child thymocytes.
    [Show full text]
  • Drosophila Pax6 Promotes Development of the Entire Eye-Antennal Disc, Thereby Ensuring Proper Adult Head Formation
    PAPER Drosophila Pax6 promotes development of the entire COLLOQUIUM eye-antennal disc, thereby ensuring proper adult head formation Jinjin Zhua, Sneha Palliyila, Chen Ranb, and Justin P. Kumara,1 aDepartment of Biology, Indiana University, Bloomington, IN 47405; and bDepartment of Biology, Stanford University, Stanford, CA 94305 Edited by Ellen V. Rothenberg, California Institute of Technology, Pasadena, CA, and accepted by Editorial Board Member Neil H. Shubin February 17, 2017 (received for review July 26, 2016) Paired box 6 (Pax6) is considered to be the master control gene for molecular battle among GRNs allows for the subdivision of the eye development in all seeing animals studied so far. In vertebrates, eye-antennal disc to be maintained within a single continuous it is required not only for lens/retina formation but also for the cellular field (13–16). Of the GRNs that are known to operate development of the CNS, olfactory system, and pancreas. Although within the eye-antennal disc, the retinal determination (RD) Pax6 plays important roles in cell differentiation, proliferation, and network, which controls eye development, is the best studied (17). patterning during the development of these systems, the underlying At the core of the RD network lie the Paired box 6 (Pax6) genes mechanism remains poorly understood. In the fruit fly, Drosophila eyeless (ey)andtwin of eyeless (toy), the SIX family member sine melanogaster, Pax6 also functions in a range of tissues, including oculis (so), the transcriptional coactivator eyes absent (eya), and the the eye and brain. In this report, we describe the function of Pax6 in Ski/Sno family member dachshund (dac)(17).
    [Show full text]
  • Role of Estrogen Receptor in Breast Cancer Cell Gene Expression
    4046 MOLECULAR MEDICINE REPORTS 13: 4046-4050, 2016 Role of estrogen receptor in breast cancer cell gene expression YABING ZHENG1, XIYING SHAO1, YUAN HUANG1, LEI SHI1, BO CHEN2, XIAOJIA WANG1, HONGJIAN YANG3, ZHANHONG CHEN1 and XIPING ZHANG3 Departments of 1Medical Oncology (Breast), 2Pathology and 3Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China Received April 28, 2015; Accepted February 23, 2016 DOI: 10.3892/mmr.2016.5018 Abstract. The aim of the present study was to establish the Europe in 2012, and the number of breast cancer-associated underlying regulatory mechanism of estrogen receptor (ER) mortalities is 131,000 (6). Furthermore, breast cancer is in breast cancer cell gene expression. A gene expression the most common cause of cancer-associated mortality in profile accession( no. GSE11324) was downloaded from the females. Therefore, it is essential to understand its molecular Gene Expression Omnibus (GEO) database. Differentially mechanism and develop more effective therapeutic methods expressed genes (DEGs) from an estrogen treatment group and for breast cancer treatment. a control group were identified. Chromatin immunoprecipita- The estrogen receptor (ER) is critical in determining the tion with high-throughput sequencing data (series GSE25710) phenotype of human breast cancers and is one of the most was obtained from the GEO for the ER binding sites, and important therapeutic targets (7). Furthermore, certain studies binding and expression target analysis was performed. A total have suggested that activation of ER is responsible for various of 3,122 DEGs were obtained and ER was demonstrated to biological processes, including cell growth and differentia- exhibit inhibition and activation roles during the regulation tion, and programmed cell death (8,9).
    [Show full text]
  • Interaction of the Human Androgen Receptor Transactivation Function with the General Transcription Factor TFIIF
    Proc. Natl. Acad. Sci. USA Vol. 94, pp. 8485–8490, August 1997 Biochemistry Interaction of the human androgen receptor transactivation function with the general transcription factor TFIIF IAIN J. MCEWAN* AND JAN-ÅKE GUSTAFSSON Department of Biosciences, Novum, Karolinska Institute, S-141 57 Huddinge, Sweden Communicated by Elwood V. Jensen, University of Hamburg, Hamburg, Germany, May 27, 1997 (received for review January 28, 1997) ABSTRACT The human androgen receptor (AR) is a tion factors, and thus the polymerase, to the promoter (re- ligand-activated transcription factor that regulates genes im- viewed in refs. 17–19). This can be achieved by direct contact portant for male sexual differentiation and development. To between the activator and the general transcription factors better understand the role of the receptor as a transcription andyor interactions by means of coactivator proteins (refs. 17 factor we have studied the mechanism of action of the N- and 19–21 and references therein). terminal transactivation function. In a protein–protein inter- In recent years a number of interactions have been described action assay the AR N terminus (amino acids 142–485) between members of the steroid–thyroid hormone receptor selectively bound to the basal transcription factors TFIIF and superfamily and basal transcription factors and co–activator the TATA-box-binding protein (TBP). Reconstitution of the proteins (see ref. 22 and references therein). However, very transactivation activity in vitro revealed that AR142–485 fused to little is known concerning the identity of interacting proteins the LexA protein DNA-binding domain was competent to with the human AR. To better understand the mechanism of activate a reporter gene in the presence of a competing DNA gene regulation by the human AR we have screened a panel of template lacking LexA binding sites.
    [Show full text]
  • DLX Genes: Roles in Development and Cancer
    cancers Review DLX Genes: Roles in Development and Cancer Yinfei Tan 1,* and Joseph R. Testa 1,2,* 1 Genomics Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, USA 2 Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA * Correspondence: [email protected] (Y.T.); [email protected] (J.R.T.) Simple Summary: DLX homeobox family genes encode transcription factors that have indispensable roles in embryonic and postnatal development. These genes are critically linked to the morphogene- sis of craniofacial structures, branchial arches, forebrain, and sensory organs. DLX genes are also involved in postnatal homeostasis, particularly hematopoiesis and, when dysregulated, oncogen- esis. DLX1/2, DLX3/4, and DLX5/6 exist as bigenes on different chromosomes, sharing intergenic enhancers between gene pairs, which allows orchestrated spatiotemporal expression. Genomic alterations of human DLX gene enhancers or coding sequences result in congenital disorders such as split-hand/foot malformation. Aberrant postnatal expression of DLX genes is associated with hematological malignancies, including leukemias and lymphomas. In several mouse models of T-cell lymphoma, Dlx5 has been shown to act as an oncogene by cooperating with activated Akt, Notch1/3, and/or Wnt to drive tumor formation. In humans, DLX5 is aberrantly expressed in lung and ovarian carcinomas and holds promise as a therapeutic target. Abstract: Homeobox genes control body patterning and cell-fate decisions during development. The homeobox genes consist of many families, only some of which have been investigated regarding a possible role in tumorigenesis. Dysregulation of HOX family genes have been widely implicated in cancer etiology.
    [Show full text]
  • New Structural Perspectives in G Protein-Coupled Receptor-Mediated Src Family Kinase Activation
    International Journal of Molecular Sciences Review New Structural Perspectives in G Protein-Coupled Receptor-Mediated Src Family Kinase Activation Sandra Berndt * and Ines Liebscher Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; [email protected] * Correspondence: [email protected]; Tel.: +49-341-9722175 Abstract: Src family kinases (SFKs) are key regulators of cell proliferation, differentiation, and survival. The expression of these non-receptor tyrosine kinases is strongly correlated with cancer development and tumor progression. Thus, this family of proteins serves as an attractive drug target. The activation of SFKs can occur via multiple signaling pathways, yet many of them are poorly understood. Here, we summarize the current knowledge on G protein-coupled receptor (GPCR)- mediated regulation of SFKs, which is of considerable interest because GPCRs are among the most widely used pharmaceutical targets. This type of activation can occur through a direct interaction between the two proteins or be allosterically regulated by arrestins and G proteins. We postulate that a rearrangement of binding motifs within the active conformation of arrestin-3 mediates Src regulation by comparison of available crystal structures. Therefore, we hypothesize a potentially different activation mechanism compared to arrestin-2. Furthermore, we discuss the probable direct regulation of SFK by GPCRs and investigate the intracellular domains of exemplary GPCRs with conserved polyproline binding motifs that might serve as scaffolding domains to allow such a direct interaction. Large intracellular domains in GPCRs are often understudied and, in general, not much Citation: Berndt, S.; Liebscher, I. is known of their contribution to different signaling pathways.
    [Show full text]
  • Expression and Function of Dlx Genes in the Osteoblast Lineage Haitao Li University of Connecticut School of Medicine and Dentistry
    University of Connecticut OpenCommons@UConn UCHC Articles - Research University of Connecticut Health Center Research 4-2008 Expression and Function of Dlx Genes in the Osteoblast Lineage Haitao Li University of Connecticut School of Medicine and Dentistry Inga Marijanovic University of Connecticut School of Medicine and Dentistry Mark S. Kronenberg University of Connecticut School of Medicine and Dentistry Ivana Erceg University of Connecticut School of Medicine and Dentistry Mary Louise Stover University of Connecticut School of Medicine and Dentistry See next page for additional authors Follow this and additional works at: https://opencommons.uconn.edu/uchcres_articles Part of the Life Sciences Commons, and the Medicine and Health Sciences Commons Recommended Citation Li, Haitao; Marijanovic, Inga; Kronenberg, Mark S.; Erceg, Ivana; Stover, Mary Louise; Velonis, Dimitrios; Mina, Mina; Upholt, William B.; Kalajzic, Ivo; and Lichtler, Alexander C., "Expression and Function of Dlx Genes in the Osteoblast Lineage" (2008). UCHC Articles - Research. 49. https://opencommons.uconn.edu/uchcres_articles/49 Authors Haitao Li, Inga Marijanovic, Mark S. Kronenberg, Ivana Erceg, Mary Louise Stover, Dimitrios Velonis, Mina Mina, William B. Upholt, Ivo Kalajzic, and Alexander C. Lichtler This article is available at OpenCommons@UConn: https://opencommons.uconn.edu/uchcres_articles/49 NIH Public Access Author Manuscript Dev Biol. Author manuscript; available in PMC 2009 May 9. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Dev Biol. 2008 April 15; 316(2): 458±470. doi:10.1016/j.ydbio.2008.01.001. Expression and Function of Dlx Genes in the Osteoblast Lineage Haitao Li1, Inga Marijanovic1, Mark S.
    [Show full text]
  • Two Independent and Interactive DNA-Binding Subdomains of the Pax6 Paired Domain Are Regulated by Alternative Splicing
    Downloaded from genesdev.cshlp.org on October 8, 2021 - Published by Cold Spring Harbor Laboratory Press Two independent and interactive DNA-binding subdomains of the Pax6 paired domain are regulated by alternative splicing Jonathan A. Epstein, 1'2 Tom Glaser, 2 Jiexing Cai, 2 Lisa Jepeal, 2 David S. Walton, 3 and Richard L. Maas 2"4 Divisions of Cardiology~ and Genetics2, Department of Medicine and Howard Hughes Medical Institute, Brigham and Women's Hospital, and Massachusetts Eye and Ear Infirmary, a Harvard Medical School, Boston, Massachusetts 02115 USA Vertebrate Pax proteins share a conserved 128-amino-acid DNA-binding motif, the paired domain. The PAX6 gene, which is mutated in the routine Small eye and human aniridia developmental defects, also encodes a second protein with a 14-amino-acid insertion in the paired domain. This protein, which arises by alternative mRNA splicing, exhibits unique DNA-binding properties. Unlike other paired domains, which bind DNA predominantly by their amino termini, the extended Pax6 paired domain interacts with DNA exclusively through its carboxyl terminus. This property can be simulated by deletion of 30 amino-terminal residues from the Pax6 or Pax2 paired domains. Thus, the insertion acts as a molecular toggle to unmask the DNA-binding potential of the carboxyl terminus. The functional nonequivalence of the two Pax6 proteins is underscored by a T ~ C mutation at position -3 of the alternative splice acceptor site that changes the ratio of the two isoforms and causes a distinct human ocular syndrome. [Key Words: Pax genes; transcription factor; alternative mRNA splicing; DNA binding; ocular development; aniridia] Received May 17, 1994; revised version accepted July 18, 1994.
    [Show full text]
  • PAX Genes in Childhood Oncogenesis: Developmental Biology Gone Awry?
    Oncogene (2015) 34, 2681–2689 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc REVIEW PAX genes in childhood oncogenesis: developmental biology gone awry? P Mahajan1, PJ Leavey1 and RL Galindo1,2,3 Childhood solid tumors often arise from embryonal-like cells, which are distinct from the epithelial cancers observed in adults, and etiologically can be considered as ‘developmental patterning gone awry’. Paired-box (PAX) genes encode a family of evolutionarily conserved transcription factors that are important regulators of cell lineage specification, migration and tissue patterning. PAX loss-of-function mutations are well known to cause potent developmental phenotypes in animal models and underlie genetic disease in humans, whereas dysregulation and/or genetic modification of PAX genes have been shown to function as critical triggers for human tumorigenesis. Consequently, exploring PAX-related pathobiology generates insights into both normal developmental biology and key molecular mechanisms that underlie pediatric cancer, which are the topics of this review. Oncogene (2015) 34, 2681–2689; doi:10.1038/onc.2014.209; published online 21 July 2014 INTRODUCTION developmental mechanisms and PAX genes in medical (adult) The developmental mechanisms necessary to generate a fully oncology. patterned, complex organism from a nascent embryo are precise. Undifferentiated primordia undergo a vast array of cell lineage specification, migration and patterning, and differentiate into an STRUCTURAL MOTIFS DEFINE THE PAX FAMILY SUBGROUPS ensemble of interdependent connective, muscle, nervous and The mammalian PAX family of transcription factors is comprised of epithelial tissues. Dysregulation of these precise developmental nine members that function as ‘master regulators’ of organo- programs cause various diseases/disorders, including—and genesis4 (Figure 1).
    [Show full text]
  • Pax Genes and Their Roles in Cell Differentiation and Development Ahmed Mansouri*, Marc Hallonetl and Peter Gruss
    851 Pax genes and their roles in cell differentiation and development Ahmed Mansouri*, Marc Hallonetl and Peter Gruss Members of the Pax gene family are expressed in various and in human aniridia [13]; in all of these, eye defects tissues during ontogenesis. Evidence for their crucial role are displayed. Pax2 is expressed during eye and kidney in morphogenesis, organogenesis, cell differentiation and development [14] and is mutated in a human family with oncogenesis is provided by rodent mutants and human kidney and eye abnormalities [15]. Pax genes clearly play diseases. Additionally, recent experimental in vivo and in vitro important roles during the formation of many structures. approaches have led to the identification of molecules that Furthermore, deregulated expression of Pax genes may interact with Pax proteins. lead to oncogenesis [16-18]. In this review, we focus on recent data documenting the Addresses role of Pax genes, and the interaction of the Pax proteins Department of Molecular Cell Biology, Max-Planck Institute for Biophysical Chemistry, Am Fassberg 11, D-37077 G/Sttingen, with other transcription factors, in the development of the Germany nervous system, in organogenesis and in cell proliferation *e-mail: [email protected] and differentiation. re-mail: [email protected] Se-mail: [email protected] Pax genes in the central nervous system Current Opinion in Cell Biology 1996, 8:851-857 The roles of Pax genes are particularly documented at © Current Biology Lid ISSN 0955-0674 the level of the nervous system, which consequently may represent a model for the study of the function of the Pax Abbreviations BMP bone morphogenetic protein genes.
    [Show full text]
  • Delta;Fosb Induction in Prefrontal Cortex by Antipsychotic
    Neuropsychopharmacology (2014) 39, 538–544 & 2014 American College of Neuropsychopharmacology. All rights reserved 0893-133X/14 www.neuropsychopharmacology.org DFosB Induction in Prefrontal Cortex by Antipsychotic Drugs is Associated with Negative Behavioral Outcomes 1,2,5 1,5 1,5 3 2 1 David M Dietz , Pamela J Kennedy , HaoSheng Sun , Ian Maze , Amy M Gancarz , Vincent Vialou , Ja Wook Koo1, Ezekiell Mouzon1, Subroto Ghose4, Carol A Tamminga4 and Eric J Nestler*,1 1 Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; 2 3 Department of Pharmacology and Toxicology and Institute on Addictions, University at Buffalo, Buffalo, NY, USA; Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY, USA; 4Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA DFosB, a FosB gene product, is induced in the prefrontal cortex (PFC) by repeated exposure to several stimuli including antipsychotic drugs such as haloperidol. However, the functional consequences of increased DFosB expression following antipsychotic treatment have not been explored. Here, we assessed whether DFosB induction by haloperidol mediates the positive or negative consequences or clinical-related actions of antipsychotic treatment. We show that individuals with schizophrenia who were medicated with antipsychotic drugs at their time of death display increased DFosB levels in the PFC, an effect that is replicated in rats treated chronically with haloperidol. In contrast, individuals with schizophrenia who were medication-free did not exhibit this effect. Viral-mediated overexpression of DFosB in the PFC of rodents induced cognitive deficits as measured by inhibitory avoidance, increased startle responses in prepulse inhibition tasks, and increased MK-801-induced anxiety-like behaviors.
    [Show full text]
  • C-Fos Assessment As a Marker of Anti–Epidermal Growth Factor Receptor Effect
    Research Article C-fos Assessment as a Marker of Anti–Epidermal Growth Factor Receptor Effect Antonio Jimeno,1 Peter Kulesza,1,2 Erik Kincaid,2 Nadia Bouaroud,1 Audrey Chan,1 Arlene Forastiere,1 Julie Brahmer,1 Douglas P. Clark,2 and Manuel Hidalgo1 1The Sidney Kimmel Comprehensive Cancer Center and 2Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland Abstract lines (1, 2). Despite the ubiquitous expression of the EGFR and the large number of patients treated in clinical trials with Factors predicting sensitivity to epidermal growth factor receptor (EGFR) blockade are largely unknown and new EGFR-targeted agents, the factors determining and predicting strategies are being sought to individualize cancer therapy. their efficacy are largely unknown. Recent reports have suggested This study evaluated the variation in the expression of the that the presence of acquired mutations in the catalytic domain of early response gene c-fos as a distal effect of EGFR inhibi- the egfr gene increase sensitivity to anti-EGFR small-molecule tion and its relationship to antitumor effects. The growth- inhibitors in non–small-cell lung cancer (3, 4); however, the inhibitory and c-fos–modulating effects of gefitinib and robustness and impact in clinical decision-making strategies of erlotinib in human cancer cell lines (A431, CAL27, HN11, those retrospective observations are currently undetermined and HuCCT1, and Hep2) were determined. Next, these cell lines more systematic reports do not support the initial observations were xenografted in mice and treated for 14 days with gefitinib (5, 6). The sole assessment of pretreatment markers may not be sufficient and even conflicting (7) and a posttreatment marker may (A431 and HuCCT1) or erlotinib (CAL27, HN11, and Hep2).
    [Show full text]