RASSF6 Is a Novel Member of the RASSF Family of Tumor Suppressors

Total Page:16

File Type:pdf, Size:1020Kb

RASSF6 Is a Novel Member of the RASSF Family of Tumor Suppressors Oncogene (2007) 26, 6203–6211 & 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00 www.nature.com/onc ORIGINAL ARTICLE RASSF6 is a novel member of the RASSF family of tumor suppressors NPC Allen1, H Donninger2, MD Vos1, K Eckfeld1, L Hesson3, L Gordon2, MJ Birrer1, F Latif3 and GJ Clark1,2 1Department of Cell and Cancer Biology, National Cancer Institute, Rockville, MD, USA; 2Molecular Targets Group, Department of Medicine, JG Brown Cancer Center, University of Louisville, Louisville, KY, USA and 3Section of Medical and Molecular Genetics, University of Birmingham, Birmingham, UK RASSF family proteins are tumor suppressors that are activated forms of Ras can also exhibit growth frequently downregulated during the development of antagonistic properties. These include the induction of human cancer. The best-characterizedmember of the senescence, cell cycle arrest and apoptosis (Mayo et al., family is RASSF1A, which is downregulated by promoter 1997; Serrano et al., 1997; Nicke et al., 2005). We can methylation in 40–90% of primary human tumors. We reconcile these apparently contradictory properties if we now identify and characterize a novel member of the consider the fact that Ras proteins can interact with a RASSF family, RASSF6. Like the other family members, wide variety of downstream effector proteins allowing a RASSF6 possesses a Ras Association domain and binds broad range of effector outputs (Malumbres and activatedRas. Exogenous expression of RASSF6 pro- Pellicer, 1998). Many Ras effector proteins contain motedapoptosis, synergizedwith activatedK-Ras to conserved structural regions that are responsible for induce cell death and inhibited the survival of specific mediating the interaction with Ras. These regions have tumor cell lines. Suppression of RASSF6 enhancedthe been designated the Ras Association (RA) domain tumorigenic phenotype of a human lung tumor cell line. (Ponting and Benjamin, 1996). Using a bioinformatics- Furthermore, RASSF6 is often downregulated in primary based approach to screen for RA domain containing human tumors. RASSF6 shares some similar overall proteins, we have previously identified several members properties as other RASSF proteins. However, there are of the RASSF family as potential Ras effectors that significant differences in biological activity between mediate growth inhibitory effects. RASSF6 and other family members including a discrete Four RASSF proteins have been characterized biolo- tissue expression profile, cell killing specificity andimpact gically thus far: RASSF1, RASSF2, RASSF4 and Nore1 on signaling pathways. Moreover, RASSF6 may play a (RASSF5) (Dammann et al., 2000; Vos et al., 2000, role in dictating the degree of inflammatory response to 2003a, b; Burbee et al., 2001; Khokhlatchev et al., 2002; the respiratory syncytial virus. Thus, RASSF6 is a novel Eckfeld et al., 2004; Agathanggelou et al., 2005). All RASSF family member that demonstrates the properties demonstrate biological properties compatible with a of a Ras effector andtumor suppressor but exhibits tumor suppressor function. These include inhibiting biological properties that are unique anddistinctfrom growth, promoting cell cycle arrest and apoptosis. They those of other family members. are all frequently downregulated during tumorigenesis by Oncogene (2007) 26, 6203–6211; doi:10.1038/sj.onc.1210440; promoter methylation (Agathanggelou et al., 2005; Akino published online 2 April 2007 et al., 2005). Moreover, RASSF1A knockout mice demon- strate enhanced tumor susceptibility (Tommasi et al., Keywords: Ras; RASSF6; tumor suppressor 2005) and loss of Nore1 (RASSF5) function is linked to a familial form of kidney cancer (Chen et al., 2003). Here, we identify and characterize a novel member of this family, RASSF6. RASSF6 interacts directly with K-Ras in a guanosine triphosphate (GTP)-dependent Introduction manner via its effector domain with an affinity compar- able to that of other known Ras effectors. RASSF6 Mutant Ras proteins play a critical role in the induces apoptosis and the ability of RASSF6 to kill development of over 30% of human cancers (Shields cells is enhanced by activated Ras. Overexpression et al., 2000; Malumbres and Barbacid, 2003). However, of RASSF6 inhibits the survival of specific tumor cell although promoting many aspects of transformation, lines and knockdown of RASSF6 by siRNA enhances the ability of tumor cell lines to grow in soft agar. In matched pair primary tumor samples, the levels Correspondence: Dr GJ Clark, JG Brown Cancer Center, Department of RASSF6 mRNA are often downregulated in the Medicine, Molecular Targets Group, University of Louisville, 580 S primary tumors. However, although RASSF6 has some Preston Street, Louisville, KY 40202, USA. E-mail: [email protected] similar overall properties as RASSF1A, the proteins Received 4 January 2007; revised 22 February 2007; accepted 22 demonstrate discrete expression profiles, and cell killing February 2007; published online 2 April 2007 specificity. RASSF6 is a potential Ras effector/tumor suppressor NPC Allen et al 6204 Recently, the RASSF6 locus has been implicated in RASSF6 and K-Ras, we generated a recombinant determining susceptibility to infection by the respiratory glutathione S-transferase (GST)-fusion of the RA domain syncytial virus (RSV) (Hull et al., 2004). RSV infection of RASSF6, designated GST-F6RA. GST-F6RA was activates the eukaryotic nuclear factor kB (NFkB) used as an affinity reagent in quantitative binding assays pathway and this activation may play a vital role in with purified, farnesylated K-Ras (Figure 3). Ras was the inflammatory response to infection. We have found loaded with GTP and incubated with 100 ng of GST or that RASSF6 expression inhibits the basal levels of GST-F6RA at decreasing concentrations for 4 h at 41Cin NFkB activity in a lung epithelial cell line, suggesting phosphate-buffered saline, 0.025% Tween-20 and 1 mM that defects in RASSF6 may facilitate viral NFkB MgCl2. GST-F6 beads were then washed and subjected to activation. Thus, we identify a further member of the western analysis using an anti-K-Ras antibody (F234 RASSF family as a novel potential Ras effector/tumor Santa Cruz Biotechnology, CA, USA). The apparent Kd suppressor with distinct biological characteristics. was taken as half the maximal binding. Binding was saturated by 300 nM,givinganapparentKd of less than or equal to 150 nM. Results Family alignment and tissue distribution of RASSF6 RASSF6 and RASSF1A exhibit different cell-specific TblastN searches of the EST database using the RA growth inhibitory properties domain of RASSF1A led to the identification of Tumor cell lines were transfected with pBabe expression RASSF6. During the preparation of this manuscript, constructs and selected in puromycin. Colony formation the gene was described in: http://www.genecards.org/ was scored after 2 weeks (Figure 4). Whereas neither cgi-bin/carddisp.pl?gene ¼ RASSF6. Sequences were RASSF1A nor RASSF6 inhibited the growth of the aligned using ClustalW (Figure 1a). RASSF6 shows H1299 human lung tumor cell line, both inhibited the most identity to RASSF2 (57%) and RASSF4 (55%). It growth of MCF-7 human breast tumor cells. However, lacks the N-terminal extension of Nore1 (RASSF5) and RASSF6 was significantly less effective at inhibiting the the microtubule association domain of RASSF1A (Vos growth and survival of A549 human lung tumor cells. et al., 2004; Agathanggelou et al., 2005) but shares the SARAH motif of other family members. Northern analysis of RASSF6 expression (Figure 1b) showed a RASSF6 and K-Ras synergize to induce cell death distinct pattern of expression that differs from other To determine the effects of activated Ras on the family members (Vos et al., 2000, 2003a, b). biological activity of RASSF6, we performed cotrans- fections in 293-T cells. After 48 h, the cells were stained RASSF6 interacts with Ras GTP in cells via its with trypan blue to detect cell death. A dramatic effector domain synergistic activation of cell death was observed when 293-T cells were cotransfected with FLAG-tagged the cells were transfected with activated K-Ras and RASSF6 and HA-tagged activated K-Ras (G12V) or RASSF6 together (Figure 5aand b). wild-type K-Ras. The cells were lysed after 48 h and immunoprecipitated with anti-Ras antibody for western RASSF6 induces apoptotic cell death analysis using anti-FLAG antibodies. RASSF6 pre- To determine if RASSF6 was proapoptotic, we used the ferentially associated with the active form of K-Ras as pCaspase3-Sensor system (Clontech, Mountain View, no association was seen with the inactive (wild-type) CA, USA) and fluorescent microscopy to measure protein (Figure 2a). caspase-3 activation in individual cells as described Ras associates with its effectors via its effector previously (Vos et al., 2003b). Quantification showed domain (Marshall 1993) therefore, effector mutants that addition of RASSF6 induced a fivefold increase of (White et al., 1995; Vos et al., 2003a) of activated active caspases over empty vector (Figure 6). K-Ras (G12V/35S) and (G12V/40C) were subjected to similar coprecipitation experiments with RASSF6. Figure 2b shows that mutations in the effector region of RASSF6 binds the proapoptotic protein MOAP-1 K-Ras strongly impaired its ability to interact with One of the proapoptotic pathways that has been RASSF6. identified for RASSF1A involves the direct interaction Farnesylation is important for Ras to bind to its with the protein modulator of apoptosis 1 (MOAP-1) effectors with
Recommended publications
  • Supplemental Table 1. Complete Gene Lists and GO Terms from Figure 3C
    Supplemental Table 1. Complete gene lists and GO terms from Figure 3C. Path 1 Genes: RP11-34P13.15, RP4-758J18.10, VWA1, CHD5, AZIN2, FOXO6, RP11-403I13.8, ARHGAP30, RGS4, LRRN2, RASSF5, SERTAD4, GJC2, RHOU, REEP1, FOXI3, SH3RF3, COL4A4, ZDHHC23, FGFR3, PPP2R2C, CTD-2031P19.4, RNF182, GRM4, PRR15, DGKI, CHMP4C, CALB1, SPAG1, KLF4, ENG, RET, GDF10, ADAMTS14, SPOCK2, MBL1P, ADAM8, LRP4-AS1, CARNS1, DGAT2, CRYAB, AP000783.1, OPCML, PLEKHG6, GDF3, EMP1, RASSF9, FAM101A, STON2, GREM1, ACTC1, CORO2B, FURIN, WFIKKN1, BAIAP3, TMC5, HS3ST4, ZFHX3, NLRP1, RASD1, CACNG4, EMILIN2, L3MBTL4, KLHL14, HMSD, RP11-849I19.1, SALL3, GADD45B, KANK3, CTC- 526N19.1, ZNF888, MMP9, BMP7, PIK3IP1, MCHR1, SYTL5, CAMK2N1, PINK1, ID3, PTPRU, MANEAL, MCOLN3, LRRC8C, NTNG1, KCNC4, RP11, 430C7.5, C1orf95, ID2-AS1, ID2, GDF7, KCNG3, RGPD8, PSD4, CCDC74B, BMPR2, KAT2B, LINC00693, ZNF654, FILIP1L, SH3TC1, CPEB2, NPFFR2, TRPC3, RP11-752L20.3, FAM198B, TLL1, CDH9, PDZD2, CHSY3, GALNT10, FOXQ1, ATXN1, ID4, COL11A2, CNR1, GTF2IP4, FZD1, PAX5, RP11-35N6.1, UNC5B, NKX1-2, FAM196A, EBF3, PRRG4, LRP4, SYT7, PLBD1, GRASP, ALX1, HIP1R, LPAR6, SLITRK6, C16orf89, RP11-491F9.1, MMP2, B3GNT9, NXPH3, TNRC6C-AS1, LDLRAD4, NOL4, SMAD7, HCN2, PDE4A, KANK2, SAMD1, EXOC3L2, IL11, EMILIN3, KCNB1, DOK5, EEF1A2, A4GALT, ADGRG2, ELF4, ABCD1 Term Count % PValue Genes regulation of pathway-restricted GDF3, SMAD7, GDF7, BMPR2, GDF10, GREM1, BMP7, LDLRAD4, SMAD protein phosphorylation 9 6.34 1.31E-08 ENG pathway-restricted SMAD protein GDF3, SMAD7, GDF7, BMPR2, GDF10, GREM1, BMP7, LDLRAD4, phosphorylation
    [Show full text]
  • Growth and Molecular Profile of Lung Cancer Cells Expressing Ectopic LKB1: Down-Regulation of the Phosphatidylinositol 3؅-Phosphate Kinase/PTEN Pathway1
    [CANCER RESEARCH 63, 1382–1388, March 15, 2003] Growth and Molecular Profile of Lung Cancer Cells Expressing Ectopic LKB1: Down-Regulation of the Phosphatidylinositol 3؅-Phosphate Kinase/PTEN Pathway1 Ana I. Jimenez, Paloma Fernandez, Orlando Dominguez, Ana Dopazo, and Montserrat Sanchez-Cespedes2 Molecular Pathology Program [A. I. J., P. F., M. S-C.], Genomics Unit [O. D.], and Microarray Analysis Unit [A. D.], Spanish National Cancer Center, 28029 Madrid, Spain ABSTRACT the cell cycle in G1 (8, 9). However, the intrinsic mechanism by which LKB1 activity is regulated in cells and how it leads to the suppression Germ-line mutations in LKB1 gene cause the Peutz-Jeghers syndrome of cell growth is still unknown. It has been proposed that growth (PJS), a genetic disease with increased risk of malignancies. Recently, suppression by LKB1 is mediated through p21 in a p53-dependent LKB1-inactivating mutations have been identified in one-third of sporadic lung adenocarcinomas, indicating that LKB1 gene inactivation is critical in mechanism (7). In addition, it has been observed that LKB1 binds to tumors other than those of the PJS syndrome. However, the in vivo brahma-related gene 1 protein (BRG1) and this interaction is required substrates of LKB1 and its role in cancer development have not been for BRG1-induced growth arrest (10). Similar to what happens in the completely elucidated. Here we show that overexpression of wild-type PJS, Lkb1 heterozygous knockout mice show gastrointestinal hamar- LKB1 protein in A549 lung adenocarcinomas cells leads to cell-growth tomatous polyposis and frequent hepatocellular carcinomas (11, 12). suppression. To examine changes in gene expression profiles subsequent to Interestingly, the hamartomas, but not the malignant tumors, arising in exogenous wild-type LKB1 in A549 cells, we used cDNA microarrays.
    [Show full text]
  • Epigenetic Inactivation of the RAS-Effector Gene RASSF2 in Lung Cancers
    169-173 30/5/07 14:58 Page 169 INTERNATIONAL JOURNAL OF ONCOLOGY 31: 169-173, 2007 169 Epigenetic inactivation of the RAS-effector gene RASSF2 in lung cancers KYOICHI KAIRA1, NORIAKI SUNAGA1, YOSHIO TOMIZAWA3, NORIKO YANAGITANI1, TAMOTSU ISHIZUKA1, RYUSEI SAITO3, TAKASHI NAKAJIMA2 and MASATOMO MORI1 Departments of 1Medicine and Molecular Science, 2Tumor Pathology, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma 371-8511; 3Department of Respiratory Medicine, National Nishigunma Hospital, 2854 Kanai, Shibukawa, Gunma 377-8511, Japan Received December 28, 2006; Accepted March 16, 2007 Abstract. RASSF2, a member of the RAS association domain Introduction family 1 (RASSF1), is a candidate tumor suppressor gene (TSG) that is silenced by promoter hypermethylation in several Recent advances in molecular genetics have revealed that human cancers. In this study, we examined the expression of multiple alterations accumulate in a multi-step manner via RASSF2 mRNA and the promoter methylation status in lung both genetic and epigenetic mechanisms during the process cancer cell lines and in tumor samples of 106 primary non- of lung carcinogenesis (1). Aberrant DNA hypermethylation small cell lung cancers (NSCLCs) by methylation-specific of CpG sites in the promoter regions of a gene is a tumor- PCR. RASSF2 expression was absent in 26% of small cell acquired epigenetic event that results in the inactivation of lung cancers (SCLCs; n=27 lines) and 50% of NSCLCs tumor suppressor genes (TSGs) (2). The aberrant promoter (n=42 lines). Promoter methylation of RASSF2 was found hypermethylation of several TSGs is frequently observed in in 18% of the SCLC cell lines (n=22) and 62% of the NSCLC lung cancers and seems to play an important role in lung cell lines (n=26), and the methylation status was tightly cancer development (3).
    [Show full text]
  • Structural Basis for Autoactivation of Human Mst2 Kinase and Its Regulation by RASSF5
    Structure Article Structural Basis for Autoactivation of Human Mst2 Kinase and Its Regulation by RASSF5 Lisheng Ni,1 Sheng Li,1,4 Jianzhong Yu,3 Jungki Min,1,5 Chad A. Brautigam,2 Diana R. Tomchick,2 Duojia Pan,3 and Xuelian Luo1,* 1Department of Pharmacology 2Department of Biophysics The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX 75390, USA 3Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 4Present address: Biologics Department, Shanghai ChemPartner Co. Ltd., Shanghai 201203, China 5Present address: Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA *Correspondence: [email protected] http://dx.doi.org/10.1016/j.str.2013.07.008 SUMMARY dor (Sav1), the NDR family kinases Lats1 and Lats2 (Lats1/2), and the adaptor protein Mob1. They form a central kinase The tumor-suppressive Hippo pathway controls cascade to transduce signals from cell-surface receptors tissue homeostasis through balancing cell prolifera- (Avruch et al., 2012; Hergovich, 2012). tion and apoptosis. Activation of the kinases Mst1 In the canonical Hippo kinase cascade, Mst1/2, in complex and Mst2 (Mst1/2) is a key upstream event in this with Sav1, phosphorylate and activate the Lats1/2-Mob1 com- pathway and remains poorly understood. Mst1/2 plexes, which then phosphorylate the transcriptional coactivator and their critical regulators RASSFs contain Yes-associated protein (YAP), a major downstream target of the Hippo pathway (Dong et al., 2007; Hao et al., 2008; Hong and Salvador/RASSF1A/Hippo (SARAH) domains that Guan, 2012; Huang et al., 2005; Zhao et al., 2007).
    [Show full text]
  • Integrative Epigenomic and Genomic Analysis of Malignant Pheochromocytoma
    EXPERIMENTAL and MOLECULAR MEDICINE, Vol. 42, No. 7, 484-502, July 2010 Integrative epigenomic and genomic analysis of malignant pheochromocytoma Johanna Sandgren1,2* Robin Andersson3*, pression examination in a malignant pheochromocy- Alvaro Rada-Iglesias3, Stefan Enroth3, toma sample. The integrated analysis of the tumor ex- Goran̈ Akerstro̊ m̈ 1, Jan P. Dumanski2, pression levels, in relation to normal adrenal medulla, Jan Komorowski3,4, Gunnar Westin1 and indicated that either histone modifications or chromo- somal alterations, or both, have great impact on the ex- Claes Wadelius2,5 pression of a substantial fraction of the genes in the in- vestigated sample. Candidate tumor suppressor 1Department of Surgical Sciences genes identified with decreased expression, a Uppsala University, Uppsala University Hospital H3K27me3 mark and/or in regions of deletion were for SE-75185 Uppsala, Sweden 2 instance TGIF1, DSC3, TNFRSF10B, RASSF2, HOXA9, Department of Genetics and Pathology Rudbeck Laboratory, Uppsala University PTPRE and CDH11. More genes were found with in- SE-75185 Uppsala, Sweden creased expression, a H3K4me3 mark, and/or in re- 3The Linnaeus Centre for Bioinformatics gions of gain. Potential oncogenes detected among Uppsala University those were GNAS, INSM1, DOK5, ETV1, RET, NTRK1, SE-751 24 Uppsala, Sweden IGF2, and the H3K27 trimethylase gene EZH2. Our ap- 4Interdisciplinary Centre for Mathematical and proach to associate histone methylations and DNA Computational Modelling copy number changes to gene expression revealed ap- Warsaw University parent impact on global gene transcription, and en- PL-02-106 Warszawa, Poland abled the identification of candidate tumor genes for 5Corresponding author: Tel, 46-18-471-40-76; further exploration.
    [Show full text]
  • STRIPAK Complexes in Cell Signaling and Cancer
    Oncogene (2016), 1–9 © 2016 Macmillan Publishers Limited All rights reserved 0950-9232/16 www.nature.com/onc REVIEW STRIPAK complexes in cell signaling and cancer Z Shi1,2, S Jiao1 and Z Zhou1,3 Striatin-interacting phosphatase and kinase (STRIPAK) complexes are striatin-centered multicomponent supramolecular structures containing both kinases and phosphatases. STRIPAK complexes are evolutionarily conserved and have critical roles in protein (de) phosphorylation. Recent studies indicate that STRIPAK complexes are emerging mediators and regulators of multiple vital signaling pathways including Hippo, MAPK (mitogen-activated protein kinase), nuclear receptor and cytoskeleton remodeling. Different types of STRIPAK complexes are extensively involved in a variety of fundamental biological processes ranging from cell growth, differentiation, proliferation and apoptosis to metabolism, immune regulation and tumorigenesis. Growing evidence correlates dysregulation of STRIPAK complexes with human diseases including cancer. In this review, we summarize the current understanding of the assembly and functions of STRIPAK complexes, with a special focus on cell signaling and cancer. Oncogene advance online publication, 15 February 2016; doi:10.1038/onc.2016.9 INTRODUCTION in the central nervous system and STRN4 is mostly abundant in Recent proteomic studies identified a group of novel multi- the brain and lung, whereas STRN3 is ubiquitously expressed in 5–9 component complexes named striatin (STRN)-interacting phos- almost all tissues. STRNs share a
    [Show full text]
  • A Systems Genetics Approach to Revealing the Pdgfb Molecular Network of the Retina
    Molecular Vision 2020; 26:459-471 <http://www.molvis.org/molvis/v26/459> © 2020 Molecular Vision Received 19 November 2019 | Accepted 17 June 2020 | Published 19 June 2020 A systems genetics approach to revealing the Pdgfb molecular network of the retina Shasha Li,1,2 Fuyi Xu,2 Lin Liu,1 Rong Ju,3 Jonas Bergquist,1,4 Qing Yin Zheng,5,6 Jia Mi,1 Lu Lu,2 Xuri Li,3 Geng Tian1 (The first two authors contributed equally to this work.) 1Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong, China; 2Department of Genetics, Genomics and informatics, University of Tennessee Health Science Center, Memphis, TN; 3State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China; 4Analytical Chemistry and Neurochemistry, Department of Chemistry-BMC, Uppsala University, Uppsala, Sweden; 5Transformative Otology and Neuroscience Center, Case Western Reserve University School of Medicine, Cleveland, OH; 6Departments of Otolaryngology, Case Western Reserve University School of Medicine, Cleveland, OH. Purpose: Platelet-derived growth factor (PDGF) signaling is well known to be involved in vascular retinopathies. Among the PDGF family, the subunit B (PDGFB) protein is considered a promising therapeutic target. This study aimed to identify the genes and potential pathways through which PDGFB affects retinal phenotypes by using a systems genetics approach. Methods: Gene expression data had been previously generated in a laboratory for the retinas of 75 C57BL/6J(B6) X DBA/2J (BXD) recombinant inbred (RI) strains. Using this data, the genetic correlation method was used to identify genes correlated to Pdgfb. A correlation between intraocular pressure (IOP) and Pdgfb was calculated based on the Pearson correlation coefficient.
    [Show full text]
  • UNIVERSITY of CALIFORNIA, SAN DIEGO Functional
    ! UNIVERSITY OF CALIFORNIA, SAN DIEGO Functional characterization of the tumor suppressor RASSF2 in Acute Myelogenous Leukemia via CRISPR/Cas9-mediation A thesis submitted in partial satisfaction of the requirements for the degree Master of Science in Biology by Michael Bao Pu Wu Committee in charge: Dong-Er Zhang, Chair Stanley Lo Yang Xu 2016 ! ! ! ! The thesis of Michael Bao Pu Wu is approved, and it is acceptable in quality and form for publication on microfilm and electronically: Chair University of California, San Diego 2016 !iii ! Dedication This thesis is dedicated to my parents: Jackson Wu and Connie Chen and to my mentors, Dr. Dong-Er Zhang and Samuel A. Stoner and finally, to my truly amazing friends. I would not have made it thus far without your leading, loving, and guiding. ! iv ! Table of Contents Signature Page……………………………………………………………………… iii Dedication…..………………………………………………………………………. iv Table of Contents…………………………………………………………………… v List of Figures……………………………………………………….……………… vi Acknowledgements….…………………………………………………………….. vii Abstract of the Thesis………………………………………………………………viii I. Introduction………………………………………………………………………... 1 II. Results…………………………………………………………………………….. 8 III. Discussion……………………………………………………………………….30 IV. Materials and Methods…………………………………………………………. 34 References…………………………………………………………………………...37 ! v ! List of Figures Figure i: Relative RASSF2 mRNA transcript expression was compared by RT-qPCR in primary CD34+ cells isolated from human cord blood, two t(8;21) AML cell lines: SKNO-1 and Kasumi-1, and a FAB subtype M2 non-t(8;21) AML cell lines: HL-60, U937, and NB4. Data are normalized to expression in primary CD34+ cell controls..4 Figure ii: Relative RASSF2 mRNA transcript expression of HL-60 transduced with MIP- RUNX1-ETO compared to non-transduced HL-60 at 48 and 72 hour timepoints.
    [Show full text]
  • RASSF2 Antibody Goat Polyclonal Antibody Catalog # ALS12987
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 RASSF2 Antibody Goat Polyclonal Antibody Catalog # ALS12987 Specification RASSF2 Antibody - Product Information Application IHC Primary Accession P50749 Reactivity Human, Rabbit, Monkey, Pig, Horse, Bovine, Dog Host Goat Clonality Polyclonal Calculated MW 38kDa KDa RASSF2 Antibody - Additional Information Gene ID 9770 Anti-RASSF2 antibody IHC of human liver. Other Names Ras association domain-containing protein 2, RASSF2, KIAA0168 Target/Specificity Human RASSF2. Reported variants represent identical protein: NP_055552.1; NP_739580.1. Reconstitution & Storage Store at -20°C. Minimize freezing and thawing. Precautions Anti-RASSF2 antibody IHC of human spleen. RASSF2 Antibody is for research use only and not for use in diagnostic or therapeutic procedures. RASSF2 Antibody - Protein Information Name RASSF2 Synonyms CENP-34 {ECO:0000303|PubMed:20813266}, K Function Potential tumor suppressor. Acts as a KRAS-specific effector protein. May promote Anti-RASSF2 antibody IHC of human thymus. apoptosis and cell cycle arrest. Stabilizes Page 1/2 10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 STK3/MST2 by protecting it from RASSF2 Antibody - Background proteasomal degradation. Potential tumor suppressor. Acts as a Cellular Location KRAS-specific effector protein. May promote Nucleus. Cytoplasm. Chromosome, apoptosis and cell cycle arrest. Stabilizes centromere, kinetochore. STK3/MST2 by protecting it from proteasomal Note=Translocates to the cytoplasm in the degradation. presence of STK3/MST2 AND STK4/MST1 RASSF2 Antibody - References Tissue Location Widely expressed with highest levels in Burbee D.G.,et al.Submitted (SEP-2002) to the brain, placenta, peripheral blood and lung.
    [Show full text]
  • The Discovery of a Novel, Ras-Mediated NORE1A/PMLIV Tumor Suppressor Complex." (2016)
    University of Louisville ThinkIR: The University of Louisville's Institutional Repository Electronic Theses and Dissertations 8-2016 The discovery of a novel, Ras-mediated NORE1A/ PMLIV tumor suppressor complex. Jessica Mezzanotte Sharpe University of Louisville Follow this and additional works at: https://ir.library.louisville.edu/etd Part of the Medical Biochemistry Commons, and the Medical Molecular Biology Commons Recommended Citation Sharpe, Jessica Mezzanotte, "The discovery of a novel, Ras-mediated NORE1A/PMLIV tumor suppressor complex." (2016). Electronic Theses and Dissertations. Paper 2539. https://doi.org/10.18297/etd/2539 This Doctoral Dissertation is brought to you for free and open access by ThinkIR: The nivU ersity of Louisville's Institutional Repository. It has been accepted for inclusion in Electronic Theses and Dissertations by an authorized administrator of ThinkIR: The nivU ersity of Louisville's Institutional Repository. This title appears here courtesy of the author, who has retained all other copyrights. For more information, please contact [email protected]. THE DISCOVERY OF A NOVEL, RAS-MEDIATED NORE1A/PMLIV TUMOR SUPPRESSOR COMPLEX By: Jessica Mezzanotte Sharpe B.A., Vanderbilt University, 2010 M.S., University of Louisville, 2014 A Dissertation Submitted to the Faculty of the School of Medicine of the University of Louisville in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy in Biochemistry and Molecular Biology Department of Biochemistry and Molecular Genetics University of Louisville Louisville, KY August, 2016 Copyright 2016 by Jessica Mezzanotte Sharpe All Rights Reserved THE DISCOVERY OF A NOVEL, RAS-MEDIATED NORE1A/PMLIV TUMOR SUPPRESSOR COMPLEX By Jessica Mezzanotte Sharpe B.A., Vanderbilt University, 2010 M.S., University of Louisville, 2014 A Dissertation Approved on June 8, 2016 by the following Dissertation Committee: __________________________________________ Dr.
    [Show full text]
  • Gene Section Short Communication
    Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Short Communication RASSF5 (Ras association (RalGDS/AF -6) domain family member 5) Lee Schmidt, Geoffrey J Clark University of Louisville, Room 119C, Baxter II Research Building, 580 S Preston Street, Louisville, KY 40202, USA (LS, GJC) Published in Atlas Database: October 2011 Online updated version : http://AtlasGeneticsOncology.org/Genes/RASSF5ID42059ch1q32.html DOI: 10.4267/2042/47279 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity DNA/RNA Other names: MGC10823, MGC17344, Maxp1, Description NORE1, NORE1A, NORE1B, RAPL, RASSF3 The human gene for RASSF5 is 81 kb in length and is HGNC (Hugo): RASSF5 located on chromosome 1(q32.1). The gene can Location: 1q32.1 produce 4 protein isoforms, two via differential exon usage, a third via differential promoter usage and the genesis of the 4 th (which can be found as an EST clone) is not yet clear. The largest isoform, A, is 418 amino acids long and has a molecular weight of about 47 kD. The protein structure of RASSF5A contains a proline- rich region at the N-terminus which contains potential SH3 binding sites and a nuclear localization signal. This is followed by a cystein rich domain, sometimes Note referred to as a zinc finger. Next is the Ras association Murine RASSF5 originally named Nore1a. Nore1B domain and this is followed by sequence containing the independently identified and designated RAPL. Rat SARAH motif required for binding to the pro-apoptotic RASSF5 also cloned independently and designated kinases MST1 and MST2.
    [Show full text]
  • The DNA Sequence and Comparative Analysis of Human Chromosome 20
    articles The DNA sequence and comparative analysis of human chromosome 20 P. Deloukas, L. H. Matthews, J. Ashurst, J. Burton, J. G. R. Gilbert, M. Jones, G. Stavrides, J. P. Almeida, A. K. Babbage, C. L. Bagguley, J. Bailey, K. F. Barlow, K. N. Bates, L. M. Beard, D. M. Beare, O. P. Beasley, C. P. Bird, S. E. Blakey, A. M. Bridgeman, A. J. Brown, D. Buck, W. Burrill, A. P. Butler, C. Carder, N. P. Carter, J. C. Chapman, M. Clamp, G. Clark, L. N. Clark, S. Y. Clark, C. M. Clee, S. Clegg, V. E. Cobley, R. E. Collier, R. Connor, N. R. Corby, A. Coulson, G. J. Coville, R. Deadman, P. Dhami, M. Dunn, A. G. Ellington, J. A. Frankland, A. Fraser, L. French, P. Garner, D. V. Grafham, C. Grif®ths, M. N. D. Grif®ths, R. Gwilliam, R. E. Hall, S. Hammond, J. L. Harley, P. D. Heath, S. Ho, J. L. Holden, P. J. Howden, E. Huckle, A. R. Hunt, S. E. Hunt, K. Jekosch, C. M. Johnson, D. Johnson, M. P. Kay, A. M. Kimberley, A. King, A. Knights, G. K. Laird, S. Lawlor, M. H. Lehvaslaiho, M. Leversha, C. Lloyd, D. M. Lloyd, J. D. Lovell, V. L. Marsh, S. L. Martin, L. J. McConnachie, K. McLay, A. A. McMurray, S. Milne, D. Mistry, M. J. F. Moore, J. C. Mullikin, T. Nickerson, K. Oliver, A. Parker, R. Patel, T. A. V. Pearce, A. I. Peck, B. J. C. T. Phillimore, S. R. Prathalingam, R. W. Plumb, H. Ramsay, C. M.
    [Show full text]