Impact of P53 Knockout and Topotecan Treatment on Gene Expression Profiles in Human Colon Carcinoma Cells: a Pharmacogenomic Study

Total Page:16

File Type:pdf, Size:1020Kb

Impact of P53 Knockout and Topotecan Treatment on Gene Expression Profiles in Human Colon Carcinoma Cells: a Pharmacogenomic Study [CANCER RESEARCH 63, 2782–2793, June 1, 2003] Impact of p53 Knockout and Topotecan Treatment on Gene Expression Profiles in Human Colon Carcinoma Cells: A Pharmacogenomic Study Sayed S. Daoud,1 Peter J. Munson, William Reinhold, Lynn Young, Vinay V. Prabhu, Qiang Yu, Jihyun LaRose, Kurt W. Kohn, John N. Weinstein, and Yves Pommier2 Department of Pharmaceutical Sciences, Washington State University, Pullman, Washington 99164-6510 [S. S. D.], and Mathematical and Statistical Computing Laboratory, Center for Information Technology [P. J. M., L. Y., V. V. P.] and Laboratory of Molecular Pharmacology, Center for Cancer Research [W. R., Q. Y., J. L., K. W. K., J. N. W., Y. P.], National Cancer Institute, NIH, Bethesda, Maryland 20892-4255 ABSTRACT through sequence-specific binding of its central domain to cis-acting elements within the promoters or introns of responsive genes. At To uncover transcriptional stress responses related to p53, we used present, more than 20 genes are known to be activated by p53, most cDNA microarrays (National Cancer Institute Oncochips comprising 6500 of them in growth arrest or apoptotic pathways (5, 6). Other promoters different genes) to characterize the gene expression profiles of wild-type p53 HCT-116 cells and an isogenic p53 knockout counterpart after treat- (many of which are viral or growth stimulatory) are repressed by p53 ment with topotecan, a specific topoisomerase I inhibitor. The use of the (7). Consequently, the downstream effects of activating p53 are com- p53 knockout cells had the advantage over p53-overexpressing systems in plex, and no single pathway mediates the full range of functions that p53 activation is mediated physiologically. RNA was extracted after of p53. low (0.1 ␮M)- and high (1 ␮M)-dose topotecan at multiple time points To analyze the p53 dependence of molecular events after DNA within the first6hoftreatment. To facilitate simultaneous study of the damage, we compared gene expression changes in a p53 wild-type p53 status and pharmacological effects on gene expression, we developed human colon carcinoma cell line, HCT-116 (p53ϩ/ϩ), with those in a novel “cross-referenced network” experimental design and used multi- an isogenic p53 knockout (p53Ϫ/Ϫ; Ref. 8) after treatment with the ple linear least squares fitting to optimize estimates of relative transcript topoisomerase I inhibitor topotecan. Topotecan (Hycamtin), a semi- levels in the network of experimental conditions. Approximately 10% of synthetic water-soluble derivative of camptothecin, is a clinically the transcripts were up- or down-regulated in response to topotecan in the p53؉/؉ cells, whereas only 1% of the transcripts changed in the p53؊/؊ useful agent. It is approved for first-line therapy of cisplatin-refractory cells, indicating that p53 has a broad effect on the transcriptional response ovarian cancer and second-line therapy of small cell lung cancer (9). to this stress. Individual transcripts and their relationships were analyzed Like other camptothecins, topotecan converts topoisomerase I into a using clustered image maps and by a novel two-dimensional analysis/ cellular poison by trapping topoisomerase I in a covalent complex visualization, gene expression map, in which each gene expression level is with DNA. The cytotoxic lesions result from breaks, generated by represented as a function of both the genotypic/phenotypic difference (i.e., collision of the complexes with DNA or RNA polymerase (10). We p53 status) and the treatment effect (i.e., of topotecan dose and time of chose to use the p53 knockout in this study, rather than an overex- exposure). Overall, drug-induced p53 activation was associated with a pressing p53 transfectant, so that the p53 expression would be phys- coherent genetic program leading to cell cycle arrest and apoptosis. We iological. identified novel p53-induced and DNA damage-induced genes (the pro- To simultaneously study the effects of p53 status and topotecan apoptotic SIVA gene and a set of transforming growth factor ␤-related genes). Genes induced independently of p53 included the antiapoptotic treatment at different concentrations and time points, we developed cFLIP gene and known stress genes related to the mitogen-activated (and present here) a new experimental design for microarray studies. protein kinase pathway and the Fos/Jun pathway. Genes that were neg- We term it a cross-referenced network (Fig. 1). The most frequently atively regulated by p53 included members of the antiapoptotic protein used design for two-color microarray experiments simply compares chaperone heat shock protein 70 family. Finally, among the p53-depend- each sample with a single internal reference sample by cohybridiza- ent genes whose expression was independent of drug treatment was tion. The network design uses internal reference samples, but it also ؉ S100A4, a small Ca2 -binding protein that has recently been implicated in provides the additional, global set of comparisons indicated in Fig. 1. p53 binding and regulation. The new experimental design and gene ex- Given this design, we were able to analyze the entire network of data pression map analysis introduced here are applicable to a wide range of by multiple regression in an appropriately weighted fashion to in- studies that encompass both treatment effects and genotypic or pheno- crease the statistical reliability of the results and conclusions in terms typic differences. of statistical consistency test. The data were then displayed using a novel visualization, GEM,3 to identify transcripts that differed in INTRODUCTION expression level in relation to p53 status and/or drug treatment. The p53 protein is a central transcription factor activated in re- sponse to a variety of cellular stresses, including DNA damage, MATERIALS AND METHODS mitotic spindle damage, heat shock, metabolic changes, hypoxia, viral infection, and oncogene activation (1, 2). p53 can induce growth arrest Cell Culture and Drug Treatment. Isogenic p53-null (p53Ϫ/Ϫ) and and apoptosis, events that prevent the survival of damaged cells. p53 wild-type (p53ϩ/ϩ) human HCT-116 colon carcinoma cells, kindly provided can also promote early senescence in response to unregulated mito- by Dr. Bert Vogelstein (8), were grown as monolayers in DMEM (Life genic signaling (3, 4). The transactivation function of p53 is mediated Technologies, Inc., Gaithersburg, MD) supplemented with 10% fetal bovine serum (Hyclone, Logan, UT). Exponentially growing cultures at 80% conflu- ence were treated for 1 h with 0.1 ␮M (LD) or 1 ␮M (HD) topotecan obtained Received 8/30/02; accepted 3/31/03. The costs of publication of this article were defrayed in part by the payment of page from the Developmental Therapeutics Program, National Cancer Institute, NIH charges. This article must therefore be hereby marked advertisement in accordance with (Bethesda, MD). Cells were then washed twice with PBS (pH 7.4) resuspended 18 U.S.C. Section 1734 solely to indicate this fact. in drug-free medium and harvested 0, 1.5, 3, or 6 h later. Total RNA was 1 To whom requests for reprints may be addressed, at Department of Pharmaceutical Sciences Cancer Prevention & Research Center Washington State University, 259 Wegner Hall, Pullman, WA 99164-6510. E-mail: [email protected]. 3 The abbreviations used are: GEM, gene expression map; TGF, transforming growth 2 To whom requests for reprints may be addressed, at Laboratory of Molecular factor; LD, low dose; HD, high dose; CIM, clustered image map; RMS, root mean square; Pharmacology Center for Cancer Research National Cancer Institute/NIH Building 37, RT-PCR, reverse transcription-PCR; PCNA, proliferating cell nuclear antigen; HSP, heat Room 5068, Bethesda, MD 20892-4255. E-mail: [email protected]. shock protein. 2782 Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2003 American Association for Cancer Research. PHARMACOGENOMIC OF p53 AND TOPOTECAN IN COLON CANCER Experimental Design. The 12 distinct cDNA preparations were as shown in Table 1. Each chip was hybridized using two cDNA preparations, each labeled with either Cy3 or Cy5. Every such comparison of preparation i with preparation j was also performed using reciprocal labeling, and the pair was replicated n (i, j) times. To compensate for any possible systematic bias caused by the chemical labeling difference between the two dyes, the experimental log-ratio, LЉ(i, j, k), and that from the reciprocal experiment, Ϫ LЉ(j, i, k), for each replicate k were averaged to give the final estimate of the log-expression ratio y(i, j) for that comparison. n͑i, j͒ Fig. 1. Experimental design of the cDNA hybridization experiments. Total RNA from ͸ ͑LЉ͑i, j, k͒ Ϫ LЉ͑ j, i, k͒͒ p53 wild-type (positive) cells [p, HCT-116 (p53ϩ/ϩ)] and p53 knockout (minus) cells [m, HCT-116 (p53Ϫ/Ϫ)] was collected at the beginning of treatment (t ϭ 0) or at the kϭ1 ϭ ϭ y͑i, j͒ ϭ (A) indicated time point (t 1.5, 3, or 6 h) after1hoftreatment with either a low (LD 0.1 2*n͑i, j͒ ␮M) or high concentration (HD ϭ 1 ␮M) of topotecan. As indicated by arrows, pairs of conditions were directly compared by labeling test samples with Cy3 or Cy5 and reference samples with Cy5 or Cy3 and by cohybridizing equal amounts of the product to a single When an observation fell below quality control cutoffs (based on the number array slide. The arrows are double-headed to indicate that hybridizations were done in of pixels in a spot and the intensity level above background), it was dropped pairs with reversal of the colors to eliminate any dye-related bias. Initially, all times and from the average. When the reciprocally labeled data were compared with doses were compared with the t ϭ 0 samples. Cell types (p, m) were then compared at t ϭ 0,3,and6hatHD.Thenetwork of comparisons was used mathematically (by replicate data for the p6LD versus p0 comparison, we noted that agreement weighted multiple linear least-squares regression) to calculate the relative expression between reciprocally labeled data were better, in general, than would be based differences between all pairs of conditions, not just those cohybridized on the same array.
Recommended publications
  • Structural Basis for High-Affinity Peptide Inhibition of P53 Interactions with MDM2 and MDMX
    Structural basis for high-affinity peptide inhibition of p53 interactions with MDM2 and MDMX Marzena Pazgiera,1, Min Liua,b,1, Guozhang Zoua, Weirong Yuana, Changqing Lia, Chong Lia, Jing Lia, Juahdi Monboa, Davide Zellaa, Sergey G. Tarasovc, and Wuyuan Lua,2 aInstitute of Human Virology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD 21201; bThe First Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Shaanxi Province 710061, China; and cStructural Biophysics Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702 Communicated by Robert C. Gallo, University of Maryland, Baltimore, MD, January 28, 2009 (received for review September 29, 2008) The oncoproteins MDM2 and MDMX negatively regulate the ac- ligase activity (10). Structurally related to MDM2, MDMX of tivity and stability of the tumor suppressor protein p53—a cellular 490-aa residues possesses domain structures arranged similarly process initiated by MDM2 and/or MDMX binding to the N- to MDM2, except that MDMX lacks ubiquitin-ligase function terminal transactivation domain of p53. MDM2 and MDMX in many (11, 12). Growing evidence supports that in unstressed cells tumors confer p53 inactivation and tumor survival, and are impor- MDM2 primarily controls p53 stability through ubiquitylation to tant molecular targets for anticancer therapy. We screened a target the tumor suppressor protein for constitutive degradation duodecimal peptide phage library against site-specifically biotin- by the proteasome (13, 14), whereas MDMX mainly functions as ylated p53-binding domains of human MDM2 and MDMX chemi- a significant p53 transcriptional antagonist independently of cally synthesized via native chemical ligation, and identified sev- MDM2 (15, 16).
    [Show full text]
  • The Ubiquitin Proteasome System and Its Involvement in Cell Death Pathways
    Cell Death and Differentiation (2010) 17, 1–3 & 2010 Macmillan Publishers Limited All rights reserved 1350-9047/10 $32.00 www.nature.com/cdd Editorial The ubiquitin proteasome system and its involvement in cell death pathways F Bernassola1, A Ciechanover2 and G Melino1,3 Cell Death and Differentiation (2010) 17, 1–3; doi:10.1038/cdd.2009.189 Following the awarding of the 2004 Nobel Prize in Chemistry Inactivation of the proteasome following caspase-mediated to Aaron Ciechanover, Avram Hershko, and Irwin A Rose cleavage may disable the proteasome, interfering with its for the discovery of ubiquitin (Ub)-mediated degradation, role in the regulation of key cellular processes and thereby Cell Death and Differentiation has drawn the attention of facilitating induction of apoptosis. The noted recent develop- its readers to the Ub Proteasome System (UPS) and its ments show how understanding of these functions is just involvement in regulating cell death pathways.1–4 The current starting to emerge. For example, why does dIAP1 associate set of reviews is an update on this theme.5–16 with multiple E2s via its RING finger? Does dIAP1 also interact From previous review articles published in Cell Death and with the E3 – the F-box protein Morgue, which is a part of an Differentiation, it was apparent that the UPS has a major SCF E3 complex? Why does dIAP1, which is an E3, have to mechanistic role in regulating cell death via modification and interact with other ligases such as the N-end rule UBR1 and degradation of key regulatory proteins involved in
    [Show full text]
  • Microrna-29B-2-5P Inhibits Cell Proliferation by Directly Targeting
    Li et al. BMC Cancer (2018) 18:681 https://doi.org/10.1186/s12885-018-4526-z RESEARCH ARTICLE Open Access MicroRNA-29b-2-5p inhibits cell proliferation by directly targeting Cbl-b in pancreatic ductal adenocarcinoma Ce Li1,2, Qian Dong3, Xiaofang Che1,2, Ling Xu1,2, Zhi Li1,2, Yibo Fan1,2, Kezuo Hou1,2, Shuo Wang1,2, Jinglei Qu1,2, Lu Xu1,2, Ti Wen1,2, Xianghong Yang4, Xiujuan Qu1,2* and Yunpeng Liu1,2* Abstract Background: MicroRNAs can be used in the prognosis of malignancies; however, their regulatory mechanisms are unknown, especially in pancreatic ductal adenocarcinoma (PDAC). Methods: In 120 PDAC specimens, miRNA levels were assessed by quantitative real time polymerase chain reaction (qRT-PCR). Then, the role of miR-29b-2-5p in cell proliferation was evaluated both in vitro (Trypan blue staining and cell cycle analysis in the two PDAC cell lines SW1990 and Capan-2) and in vivo using a xenograft mouse model. Next, bioinformatics methods, a luciferase reporter assay, Western blot, and immunohistochemistry (IHC) were applied to assess the biological effects of Cbl-b inhibition by miR-29b-2-5p. Moreover, the relationship between Cbl-b and p53 was evaluated by immunoprecipitation (IP), Western blot, and immunofluorescence. Results: From the 120 PDAC patients who underwent surgical resection, ten patients with longest survival and ten with shortest survival were selected. We found that high miR-29b-2-5p expression was associated with good prognosis (p = 0.02). The validation cohort confirmed miR-29b-2-5p as an independent prognostic factor in PDAC (n = 100, 95% CI = 0.305–0.756, p = 0.002).
    [Show full text]
  • MSD® Ubiquitinated MDM2 Assay Whole Cell Lysate
    ® MSD Ubiquitinated MDM2 Assay Whole Cell Lysate Kit For quantitative determination in human, mouse, and rat whole cell lysate samples Alzheimer’s Disease MDM2 BioProcess Cardiac Cell Signaling Clinical Immunology Cytokines Hypoxia Immunogenicity Inflammation Metabolic Oncology Toxicology Vascular MDM2 (murine double minute 2), an E3 ubiquitin ligase and a negative regulator of p53, is a 56 kDa oncoprotein which is ubiquitinated and phosphorylated. MDM2 contains an amino terminal p53 interaction domain, an acidic domain in the region of amino acids 250–300 (phosphorylation in this region is believed to play a role in MDM2 regulation), and a carboxy-terminal RING domain Catalog Numbers 1 containing a Cis2-His2-Cis4 consensus motif which binds zinc and is responsible for the E3 ubiquitin ligase activity of MDM2. MDM2 Ubiquitinated MDM2 Whole degradation is controlled by self-ubiquitination, phosphorylation, and potentially through ubiquitination by other, not yet identified, E3 2 3 Cell Lysate Kit ligases. DNA damage and cellular stress trigger MDM2 degradation, releasing p53 from MDM2-mediated negative regulation. 4 Kit size Deletion of MDM2 in mouse models is lethal in a p53 dependent manner, and overexpression of MDM2 is seen in many cancers with 5 1 plate K152FJD-1 non-mutated p53 leading to the conclusion that MDM2 is oncogenic by way of p53 inactivation. Because of the important role p53 5 plates K152FJD-2 tumor suppression plays in many different forms of cancer, there has been extensive research on the interactions between MDM2 and 20 plates K152FJD-3 p53 and considerable interest in identifying drugs capable of modulating the MDM2‒p53 interaction.
    [Show full text]
  • Chemotherapy Induces NEDP1-Mediated Destabilization of MDM2
    Oncogene (2010) 29, 297–304 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc SHORT COMMUNICATION Chemotherapy induces NEDP1-mediated destabilization of MDM2 IR Watson1,2,BKLi1,2, O Roche1, A Blanch2, M Ohh1 and MS Irwin1,2,3 1Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; 2Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada and 3Department of Paediatrics and Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada MDM2 is an E3 ligase that promotes ubiquitin-mediated In response to DNA damage, p53 becomes phos- destruction of p53. Cellular stresses such as DNA damage phorylated by several kinases within the MDM2- can lead to p53 activation due in part to MDM2 binding domain, which prevents MDM2–p53 interac- destabilization. Here, we show that the stability of tion (Bode and Dong, 2004). The stabilization of p53 MDM2 is regulated by an ubiquitin-like NEDD8 pathway then leads to DNA repair, cell cycle arrest, senescence or and identify NEDP1 as a chemotherapy-induced isopepti- apoptosis. Recent studies have shown that MDM2 is dase that deneddylates MDM2, resulting in MDM2 destabilized in response to DNA damage, which promotes destabilization concomitant with p53 activation. Concor- p53 activation (Stommel and Wahl, 2004; Meulmeester dantly, RNAi-mediated knockdown of endogenous et al., 2005). NEDD8 is a ubiquitin-like protein that NEDP1 blocked diminution of MDM2 levels and regulates protein function through covalent modification increased chemoresistance of tumor cells. These findings of substrates such as Cullins, BCA3, EGFR, ribosomal unveil the regulation of MDM2 stability through NEDP1 L11 protein, VHL, p73 and p53 (Xirodimas, 2008).
    [Show full text]
  • Mdm2-Mediated Ubiquitylation: P53 and Beyond
    Cell Death and Differentiation (2010) 17, 93–102 & 2010 Macmillan Publishers Limited All rights reserved 1350-9047/10 $32.00 www.nature.com/cdd Review Mdm2-mediated ubiquitylation: p53 and beyond J-C Marine*,1 and G Lozano2 The really interesting genes (RING)-finger-containing oncoprotein, Mdm2, is a promising drug target for cancer therapy. A key Mdm2 function is to promote ubiquitylation and proteasomal-dependent degradation of the tumor suppressor protein p53. Recent reports provide novel important insights into Mdm2-mediated regulation of p53 and how the physical and functional interactions between these two proteins are regulated. Moreover, a p53-independent role of Mdm2 has recently been confirmed by genetic data. These advances and their potential implications for the development of new cancer therapeutic strategies form the focus of this review. Cell Death and Differentiation (2010) 17, 93–102; doi:10.1038/cdd.2009.68; published online 5 June 2009 Mdm2 is a key regulator of a variety of fundamental cellular has also emerged from recent genetic studies. These processes and a very promising drug target for cancer advances and their potential implications for the development therapy. It belongs to a large family of (really interesting of new cancer therapeutic strategies form the focus of this gene) RING-finger-containing proteins and, as most of its review. For a more detailed discussion of Mdm2 and its other members, Mdm2 functions mainly, if not exclusively, as various functions an interested reader should also consult an E3 ligase.1 It targets various substrates for mono- and/or references9–12. poly-ubiquitylation thereby regulating their activities; for instance by controlling their localization, and/or levels by The p53–Mdm2 Regulatory Feedback Loop proteasome-dependent degradation.
    [Show full text]
  • Hsp27 Silencing Coordinately Inhibits Proliferation and Promotes Fas-Induced Apoptosis by Regulating the PEA-15 Molecular Switch
    Cell Death and Differentiation (2012) 19, 990–1002 & 2012 Macmillan Publishers Limited All rights reserved 1350-9047/12 www.nature.com/cdd Hsp27 silencing coordinately inhibits proliferation and promotes Fas-induced apoptosis by regulating the PEA-15 molecular switch N Hayashi1, JW Peacock1, E Beraldi1, A Zoubeidi1,2, ME Gleave1,2 and CJ Ong*,1,3 Heat shock protein 27 (Hsp27) is emerging as a promising therapeutic target for treatment of various cancers. Although the role of Hsp27 in protection from stress-induced intrinsic cell death has been relatively well studied, its role in Fas (death domain containing member of the tumor necrosis factor receptor superfamily)-induced apoptosis and cell proliferation remains underappreciated. Here, we show that Hsp27 silencing induces dual coordinated effects, resulting in inhibition of cell proliferation and sensitization of cells to Fas-induced apoptosis through regulation of PEA-15 (15-kDa phospho-enriched protein in astrocytes). We demonstrate that Hsp27 silencing suppresses proliferation by causing PEA-15 to bind and sequester extracellular signal-regulated kinase (ERK), resulting in reduced translocation of ERK to the nucleus. Concurrently, Hsp27 silencing promotes Fas-induced apoptosis by inducing PEA-15 to release Fas-associating protein with a novel death domain (FADD), thus allowing FADD to participate in death receptor signaling. Conversely, Hsp27 overexpression promotes cell proliferation and suppresses Fas-induced apoptosis. Furthermore, we show that Hsp27 regulation of PEA-15 activity occurs in an Akt-dependent manner. Significantly, Hsp27 silencing in a panel of phosphatase and tensin homolog on chromosome 10 (PTEN) wild-type or null cell lines, and in LNCaP cells that inducibly express PTEN, resulted in selective growth inhibition of PTEN-deficient cancer cells.
    [Show full text]
  • Acute HSF1 Depletion Induces Cellular Senescence Through the MDM2
    © 2018. Published by The Company of Biologists Ltd | Journal of Cell Science (2018) 131, jcs210724. doi:10.1242/jcs.210724 RESEARCH ARTICLE Acute HSF1 depletion induces cellular senescence through the MDM2-p53-p21 pathway in human diploid fibroblasts Tsukasa Oda1, Takayuki Sekimoto1, Kiminori Kurashima1, Mitsuaki Fujimoto2, Akira Nakai2 and Takayuki Yamashita1,* ABSTRACT of tumor suppressor pathways (Campisi, 2013; He and Heat shock transcription factor 1 (HSF1) regulates the expression of a Sharpless, 2017; Muñoz-Espín and Serrano, 2014). In addition, wide array of genes, controls the expression of heat shock proteins increasing evidence indicates that senescent cells contribute (HSPs) as well as cell growth. Although acute depletion of HSF1 to the expression of various aging-related phenotypes (Baker induces cellular senescence, the underlying mechanisms are et al., 2016, 2011). These effects are mainly mediated by poorly understood. Here, we report that HSF1 depletion-induced growth arrest of tissue progenitor cells and paracrine actions senescence (HDIS) of human diploid fibroblasts (HDFs) was of SASP. independent of HSP-mediated proteostasis but dependent on Heat shock transcription factor 1 (HSF1) plays a central role in activation of the p53-p21 pathway, partly because of the increased protein homeostasis by transcriptionally activating the expression of expression of dehydrogenase/reductase 2 (DHRS2), a putative heat shock proteins (HSPs) (Anckar and Sistonen, 2011; Gomez- MDM2 inhibitor. We observed that HDIS occurred without Pastor et al., 2017; Labbadia and Morimoto, 2015). Additionally, decreased levels of major HSPs or increased proteotoxic stress HSF1 regulates the expression of various genes encoding important in HDFs. Additionally, VER155008, an inhibitor of HSP70 family regulators of cell growth, survival and metabolism (Hahn et al., proteins, increased proteotoxicity and suppressed cell growth but 2004; Mendillo et al., 2012; Takii et al., 2015).
    [Show full text]
  • Limiting the Power of P53 Through the Ubiquitin Proteasome Pathway
    Downloaded from genesdev.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW Limiting the power of p53 through the ubiquitin proteasome pathway Vinod Pant and Guillermina Lozano Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA The ubiquitin proteasome pathway is critical in restrain- Modification of p53 by ubiquitination and deubiquitina- ing the activities of the p53 tumor suppressor. Numerous tion is an important reversible mechanism that effectively E3 and E4 ligases regulate p53 levels. Additionally, regulates its functions (for reviews, see Jain and Barton deubquitinating enzymes that modify p53 directly or 2010; Brooks and Gu 2011; Love and Grossman 2012; indirectly also impact p53 function. When alterations of Hock and Vousden 2014). Mono- or polyubiquitination these proteins result in increased p53 activity, cells arrest of p53 by different E3 ligases regulates its nuclear ex- in the cell cycle, senesce, or apoptose. On the other hand, port, mitochondrial translocation, protein stability, and alterations that result in decreased p53 levels yield transcriptional activity. Another set of enzymes called tumor-prone phenotypes. This review focuses on the deubiquitinases (DUBs) can reverse these effects. Here, physiological relevance of these important regulators of we focus on ubiquitination as a mechanism for regulating p53 and their therapeutic implications. p53 stability and function and review current findings from in vivo models that evaluate the importance of the ubiquitin proteasome system in regulating p53. The p53 tumor suppressor maintains genomic integrity by primarily functioning as a sequence-specific DNA-binding Ubiquitination is critical for regulating p53 transcription factor (Vousden and Prives 2009).
    [Show full text]
  • MDM2 Amplified Sarcomas: a Literature Review
    diagnostics Review MDM2 Amplified Sarcomas: A Literature Review Raf Sciot Department of Pathology, University Hospital, University of Leuven, 3000 Leuven, Belgium; [email protected] Abstract: Murine Double Minute Clone 2, located at 12q15, is an oncogene that codes for an oncopro- tein of which the association with p53 was discovered 30 years ago. The most important function of MDM2 is to control p53 activity; it is in fact the best documented negative regulator of p53. Mutations of the tumor suppressor gene p53 represent the most frequent genetic change in human cancers. By overexpressing MDM2, cancer cells have another means to block p53. The sarcomas in which MDM2 amplification is a hallmark are well-differentiated liposarcoma/atypical lipomatous tumor, dedifferentiated liposarcoma, intimal sarcoma, and low-grade osteosarcoma. The purpose of this review is to summarize the typical clinical, histopathological, immunohistochemical, and genetic features of these tumors. Keywords: MDM2 amplification; well-differentiated liposarcoma/atypical lipomatous tumor; dedif- ferentiated liposarcoma; intimal sarcoma low grade osteosarcoma 1. Introduction Murine Double Minute Clone 2 is an oncogene, the function of which was first described in DNA associated with paired acentric chromatin bodies, termed double minutes, harbored in spontaneously transformed mouse 3T3 fibroblasts [1]. MDM2 (located at 12q15) codes for Citation: Sciot, R. MDM2 Amplified an oncoprotein of which the association with p53 was discovered 30 years ago. The protein Sarcomas: A Literature Review. functions as an E3 ubiquitin ligase that targets the p53 protein for proteasomal degradation. Diagnostics 2021, 11, 496. https:// Thus, the most important function of MDM2 is to control p53 activity; it is in fact the best doi.org/10.3390/diagnostics11030496 documented negative regulator of p53.
    [Show full text]
  • Cdc53/Cullin and the Essential Hrt1 RING–H2 Subunit of SCF Define a Ubiquitin Ligase Module That Activates the E2 Enzyme Cdc34
    Downloaded from genesdev.cshlp.org on September 27, 2021 - Published by Cold Spring Harbor Laboratory Press Cdc53/cullin and the essential Hrt1 RING–H2 subunit of SCF define a ubiquitin ligase module that activates the E2 enzyme Cdc34 Jae Hong Seol,1,5 R.M. Renny Feldman,1,5 Wolfgang Zachariae,2,5 Anna Shevchenko,3 Craig C. Correll,1 Svetlana Lyapina,1 Yong Chi,1 Marta Galova,2 Jonathan Claypool,4 Suzanne Sandmeyer,4 Kim Nasmyth,2 Andrej Shevchenko,3 and Raymond J. Deshaies1,6 1Division of Biology, California Institute of Technology, Pasadena, California 91125 USA; 2Research Institute of Molecular Pathology, A-1030 Vienna, Austria; 3Peptide and Protein Group European Molecular Biology Laboratory, 69012 Heidelberg, Germany; 4Department of Microbiology and Molecular Genetics, College of Medicine, University of California, Irvine, California 92697-4025 USA SCFCdc4 (Skp1, Cdc53/cullin, F-box protein) defines a family of modular ubiquitin ligases (E3s) that regulate diverse processes including cell cycle, immune response, and development. Mass spectrometric analysis of proteins copurifying with Cdc53 identified the RING–H2 finger protein Hrt1 as a subunit of SCF. Hrt1 shows striking similarity to the Apc11 subunit of anaphase-promoting complex. Conditional inactivation of hrt1ts Cdc4 results in stabilization of the SCF substrates Sic1 and Cln2 and cell cycle arrest at G1/S. Hrt1 assembles into recombinant SCF complexes and individually binds Cdc4, Cdc53 and Cdc34, but not Skp1. Hrt1 stimulates the E3 activity of recombinant SCF potently and enables the reconstitution of Cln2 ubiquitination by recombinant SCFGrr1. Surprisingly, SCF and the Cdc53/Hrt1 subcomplex activate autoubiquitination of Cdc34 E2 enzyme by a mechanism that does not appear to require a reactive thiol.
    [Show full text]
  • Between Genotype and Phenotype: Protein Chaperones and Evolvability
    REVIEWS BETWEEN GENOTYPE AND PHENOTYPE: PROTEIN CHAPERONES AND EVOLVABILITY Suzanne L. Rutherford Protein chaperones direct the folding of polypeptides into functional proteins, facilitate developmental signalling and, as heat-shock proteins (HSPs), can be indispensable for survival in unpredictable environments. Recent work shows that the main HSP chaperone families also buffer phenotypic variation. Chaperones can do this either directly through masking the phenotypic effects of mutant polypeptides by allowing their correct folding, or indirectly through buffering the expression of morphogenic variation in threshold traits by regulating signal transduction. Environmentally sensitive chaperone functions in protein folding and signal transduction have different potential consequences for the evolution of populations and lineages under selection in changing environments. CHAPERONES The heat-shock proteins (HSPs) are highly conserved proteins compromises the ability of the chaperones to A class of proteins that, families of enzymes and CHAPERONES that are involved protect or fold their usual targets, thereby reducing the by preventing improper in the folding and degradation of damaged proteins. activities of most target proteins9,10. According to recent associations, assist in the They are rapidly and concertedly mobilized in large studies, the modulation of chaperone and target func- correct folding or assembly of other proteins in vivo, but that numbers by cells that are under stress. The mobiliza- tions in response to stress would alternately mask and are not a part of the mature tion of HSPs is an important component of a univer- expose phenotypic variation, depending on the degree structure. sal and tightly orchestrated stress response that has of stress and the availability of free chaperones11–14.
    [Show full text]