Suppression of 19S Proteasome Subunits Marks Emergence of an Altered Cell State in Diverse Cancers

Total Page:16

File Type:pdf, Size:1020Kb

Suppression of 19S Proteasome Subunits Marks Emergence of an Altered Cell State in Diverse Cancers Suppression of 19S proteasome subunits marks emergence of an altered cell state in diverse cancers Peter Tsvetkova,1, Ethan Sokola,b, Dexter Jina,b, Zarina Brunea, Prathapan Thirua, Mahmoud Ghandic, Levi A. Garrawayc,d,e, Piyush B. Guptaa,b,f,g, Sandro Santagatah, Luke Whitesella, and Susan Lindquista,b,i,2 aWhitehead Institute for Biomedical Research, Cambridge, MA 02142; bDepartment of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139; cBroad Institute, Cambridge, MA 02142; dDana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215; eDepartment of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115; fKoch Institute for Integrative Cancer Research, Cambridge, MA 02139; gHarvard Stem Cell Institute, Cambridge, MA 02138; hDepartment of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115; and iHoward Hughes Medical Institute, Cambridge, MA 02139 Edited by Carol Prives, Columbia University, New York, NY, and approved December 5, 2016 (received for review November 22, 2016) The use of proteasome inhibitors to target cancer’s dependence on proteasome inhibition. In this work we examine if such a mecha- altered protein homeostasis has been greatly limited by intrinsic nism is implemented by different cancers. and acquired resistance. Analyzing data from thousands of cancer lines and tumors, we find that those with suppressed expression Results of one or more 19S proteasome subunits show intrinsic protea- Sharply Reduced Expression of One 19S Proteasome Complex Subunit some inhibitor resistance. Moreover, such proteasome subunit sup- Occurs Frequently Across Diverse Cancer Cell Lines. We first char- pression is associated with poor outcome in myeloma patients, acterized relative mRNA expression levels for all genes that where proteasome inhibitors are a mainstay of treatment. Beyond encode proteasome subunits using the Genomics of Drug Sen- conferring resistance to proteasome inhibitors, proteasome subunit sitivity in Cancer (GDSC) database (20). As expected, across 789 suppression also serves as a sentinel of a more global remodeling of cancer cell lines, the average expression level of 20S subunits was highly correlated with the average expression level of 19S sub- the transcriptome. This remodeling produces a distinct gene signa- A ture and new vulnerabilities to the proapoptotic drug, ABT-263. This units (Fig. 1 ). However, in some cell lines, the expression of a frequent, naturally arising imbalance in 19S regulatory complex specific 19S subunit was much lower than the average expression of that subunit across all cell lines. To quantify this phenomenon composition is achieved through a variety of mechanisms, including MEDICAL SCIENCES for each cell line in the GDSC and the Cancer Cell Line Ency- DNA methylation, and marks the emergence of a heritably altered clopedia (CCLE) datasets, we calculated the SD from the mean and therapeutically relevant state in diverse cancers. of every 19S subunit in every cell line. Each cell line was then assigned a sigma score that is indicative of the greatest deviation drug resistance | epigenetic gene regulation | apoptosis | EMT | from the mean of any one 19S subunit in that cell line. Thus, a bortezomib sigma score of 3 indicates a cell line in which the mRNA ex- pression of at least one subunit of the 19S proteasome complex ells rely on the proteasome machinery to mediate protein was reduced by 3 SDs from the mean mRNA level of that par- Cturnover and maintain protein homeostasis (1–3). Upon ticular subunit across all cell lines. A sigma score of 2 indicates a oncogenic transformation, a myriad of proteotoxic stresses that reduction in the mRNA of 2 SDs, and so on. In 59 cancer cell lines, the mRNA level of at least one 19S tax the cellular machinery responsible for protein homeostasis σ are introduced (2, 4). These pressures cause cancer cells to rely subunit was 3 or more SDs (3 ) from the mean for that subunit heavily on enhanced proteasome function (2, 5–8). This de- pendency can be exploited using natural and synthetic com- Significance pounds that inhibit, with exquisite potency and selectivity, the catalytic function of the 20S proteasome (9, 10). Indeed, these In previous work, we used genome-wide screening to uncover a compounds are highly effective at inhibiting the growth of a wide counterintuitive mechanism by which cells can acquire resistance variety of cancer cell lines in culture. Unfortunately, their clinical to inhibitors of the proteasome’s catalytic core through experi- utility has been surprisingly limited, with therapeutic benefits mentally induced imbalances in the composition of its regulatory commonly observed for only a few types of cancer (9, 10). particle. However, in many cases, mechanisms uncovered in vitro The limited role for proteasome inhibitors as clinical chemo- for acquired resistance often do not translate to the context therapeutics can be attributed to biological processes that pro- of actual clinical cancers. Here, we show that this mechanism is mote intrinsic and acquired resistance. For example, cell culture actually deployed spontaneously and naturally in diverse human models of acquired resistance often accumulate mutations in the cancer lines and is associated not only with increased resistance catalytic subunits of the 20S proteasome (11, 12). In terms of to proteasome inhibitors both in vitro and in the clinic but clinical relevance, however, such mutations have yet to be de- also is symptomatic of a much more broadly altered state with tected in clinical samples (13). Additional mechanisms of re- a unique gene signature and drug targetable vulnerabilities. sistance include alteration of specific cellular pathways such as κ constitutive activation of NF- B (14), activation of the chaper- Author contributions: P. Tsvetkov designed research; P.B.G., L.W., and S.L. provided sci- one machinery (15, 16), or alterations in the EGFR/JAK1/ entific supervision; P. Tsvetkov and Z.B. performed research; M.G. and L.A.G. contributed STAT3 pathway (17). new reagents/analytic tools; P. Tsvetkov, E.S., D.J., P. Thiru, M.G., and P.B.G. analyzed To further explore and identify the cellular mechanisms of re- data; and P. Tsvetkov, S.S., L.W., and S.L. wrote the paper. sistance developed in the presence of distinct proteasome inhibi- Conflict of interest statement: L.A.G. is a consultant for Foundation Medicine, Novartis, tors, our group and others recently applied genome-wide genetic Boehringer Ingelheim, and Third Rock; an equity holder in Foundation Medicine; and a screens to various cell-line models (18, 19). In all models exam- member of the Scientific Advisory Board at Warp Drive. L.A.G. receives sponsored research ined, experimentally reducing the expression of one of the many support from Novartis, Astellas, BMS, and Merck. different subunits composing the 19S regulatory complex increases This article is a PNAS Direct Submission. resistance to inhibitors of the proteasome’s catalytic core (18, 19) 1To whom correspondence should be addressed. Email: [email protected]. and may show clinical relevance in multiple myeloma patients 2Deceased October 27, 2016. (18). These data suggested that transient, nonmutational mecha- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. nisms may play a key role in the ability of cancer cells to withstand 1073/pnas.1619067114/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1619067114 PNAS Early Edition | 1of6 Downloaded by guest on September 26, 2021 A GDSC dataset B 19S regulatory complex subunits (GDSC dataset) 30 r=0.84 15 p<0.0001 20 10 10 0 (Z-score sum) 20S subunits -10 Expression -20 5 > 3 SD reduction -30 -40 -20 0 20 40 4 8 0 3 PSMC1PSMC2PSMC3PSMC4PSMC5PSMC6PSMD1PSMD2PSMD3PSMD PSMD5PSMD6PSMD7PSMD PSMD9 19S subunits PSMD1PSMD11PSMD12PSMD1PSMD14ADRM1 (Z-score sum) C D 19S regulatory complex subunits (CCLE dataset) TCGA dataset 15 120 90 10 60 30 Expression 5 # of events 0 > 3 SD reduction 0 0 4 PSMD5 PSMD1 PSMC6 PSMD6 PSMD2 PSMC2 PSMC3 PSMD7 PSMD3 PSMD9 PSMC5 PSMC4 PSMD4 PSMD8 PSMC1 ADRM1 PSMD10 PSMD12 PSMD14 PSMD11 PSMD13 PSMC2PSMC3PSMC4PSMC5PSMD1PSMD2PSMD3PSMD4PSMD5PSMD6PSMD7PSMD8PSMD9 PSMD1PSMD11PSMD12PSMD13PSMD1ADRM1 Fig. 1. Reduced expression of a single 19S proteasome complex subunit occurs naturally in many cancer cells. (A) The overall expression of all 26S proteasome subunits is tightly correlated. The correlation between the z-score sum of expression of 20S proteasome subunits versus the 19S proteasome subunits is plotted across 789 cancer cell lines taken from the GDSC dataset. Spearman correlation: r = 0.84 (P < 0.0001). (B and C) Representation of the expression levels for individual 19S subunits for each cancer cell line in the GDSC (B) and the CCLE datasets (C). Red dots indicate cells that exhibit reduced expression of a given subunit by more than 3 SDs from the mean (3σ). Each dot represents an individual cell line. (D) Analysis of primary tumor expression profiles taken from TCGA dataset. The value of each bar is the number of primary tumors exhibiting a 3σ drop in expression of at least one 19S subunit. across all 789 cell lines in the GDSC dataset. For ease of ref- paragangliomas, acute myeloid leukemias, renal cell carcinomas, erence, we designated these as “3σ lines” (Fig. 1B, red dots, and and cutaneous melanomas (Fig. S1D). Dataset S1). We also analyzed the CCLE dataset that includes Interestingly, as true for the GDSC and CCLE datasets, PSMD5 mRNA expression and DNA copy-number data for 990 cell lines. was the most commonly suppressed 19S subunit gene in human Of these, 6.3% were 3σ lines, a percentage similar to that in the tumor resection samples (Fig. 1D). In addition, other 19S subunit GDSC dataset (Fig. 1C, red dots, and Dataset S1). To determine genes that commonly showed changes in the GDSC and CCLE cell- PSMD10 PSMD1 PSMC6 PSMD6 whether this strong deviation from the mean of 19S subunits line datasets such as , , ,and were D could be explained by random variation in the expression of any suppressed in tumor resection samples as well (Fig.
Recommended publications
  • The Potential Role of Necroptosis in Clinical Diseases (Review)
    INTERNATIONAL JOURNAL OF MOleCular meDICine 47: 89, 2021 The potential role of necroptosis in clinical diseases (Review) WENLI DAI1*, JIN CHENG1*, XI LENG2, XIAOQING HU1 and YINGFANG AO1 1Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing 100191; 2Medical Imaging Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China Received November 19, 2020; Accepted March 8, 2021 DOI: 10.3892/ijmm.2021.4922 Abstract. As an important type of programmed cell death in 1. Introduction addition to apoptosis, necroptosis occurs in a variety of patho‑ physiological processes, including infections, liver diseases, Necroptosis, an emerging field closely related to apoptosis, is a kidney injury, neurodegenerative diseases, cardiovascular non‑caspase‑dependent cell death that has been implicated in diseases, and human tumors. It can be triggered by a variety the pathological processes of various diseases. It is regulated of factors, such as tumor necrosis factor receptor and Toll‑like by various genes that cause regular and ordered cell death. receptor families, intracellular DNA and RNA sensors, and Through activating specific death signaling pathways, it shares interferon, and is mainly mediated by receptor‑interacting typical characteristics of necrosis, including loss of metabolic protein kinase 1 (RIP1), RIP3, and mixed lineage kinase function and subcellular changes (1,2). Receptor‑interacting domain‑like protein. A better understanding of the mechanism protein kinase 1 (RIP1) was the first signaling molecule of necroptosis may be useful in the development of novel drugs identified in the necrosome (3). RIP1 and RIP3 interact for necroptosis‑related diseases.
    [Show full text]
  • Gene Essentiality Landscape and Druggable Oncogenic Dependencies in Herpesviral Primary Effusion Lymphoma
    ARTICLE DOI: 10.1038/s41467-018-05506-9 OPEN Gene essentiality landscape and druggable oncogenic dependencies in herpesviral primary effusion lymphoma Mark Manzano1, Ajinkya Patil1, Alexander Waldrop2, Sandeep S. Dave2, Amir Behdad3 & Eva Gottwein1 Primary effusion lymphoma (PEL) is caused by Kaposi’s sarcoma-associated herpesvirus. Our understanding of PEL is poor and therefore treatment strategies are lacking. To address this 1234567890():,; need, we conducted genome-wide CRISPR/Cas9 knockout screens in eight PEL cell lines. Integration with data from unrelated cancers identifies 210 genes as PEL-specific oncogenic dependencies. Genetic requirements of PEL cell lines are largely independent of Epstein-Barr virus co-infection. Genes of the NF-κB pathway are individually non-essential. Instead, we demonstrate requirements for IRF4 and MDM2. PEL cell lines depend on cellular cyclin D2 and c-FLIP despite expression of viral homologs. Moreover, PEL cell lines are addicted to high levels of MCL1 expression, which are also evident in PEL tumors. Strong dependencies on cyclin D2 and MCL1 render PEL cell lines highly sensitive to palbociclib and S63845. In summary, this work comprehensively identifies genetic dependencies in PEL cell lines and identifies novel strategies for therapeutic intervention. 1 Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA. 2 Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA. 3 Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA. Correspondence and requests for materials should be addressed to E.G. (email: [email protected]) NATURE COMMUNICATIONS | (2018) 9:3263 | DOI: 10.1038/s41467-018-05506-9 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-05506-9 he human oncogenic γ-herpesvirus Kaposi’s sarcoma- (IRF4), a critical oncogene in multiple myeloma33.
    [Show full text]
  • Large-Scale Image-Based Profiling of Single-Cell Phenotypes in Arrayed CRISPR-Cas9 Gene Perturbation Screens
    Published online: January 23, 2018 Method Large-scale image-based profiling of single-cell phenotypes in arrayed CRISPR-Cas9 gene perturbation screens Reinoud de Groot1 , Joel Lüthi1,2 , Helen Lindsay1 , René Holtackers1 & Lucas Pelkmans1,* Abstract this reason, CRISPR-Cas9 has been used in large-scale functional genomic screens (Shalem et al, 2015). Most screens performed to High-content imaging using automated microscopy and computer date employ a pooled screening strategy, which can identify genes vision allows multivariate profiling of single-cell phenotypes. Here, that cause differential growth in screening conditions (Koike-Yusa we present methods for the application of the CISPR-Cas9 system et al, 2013; Shalem et al, 2014; Wang et al, 2014). However, in large-scale, image-based, gene perturbation experiments. We pooled screening precludes multivariate profiling of single-cell show that CRISPR-Cas9-mediated gene perturbation can be phenotypes. This can be partially overcome by combining pooled achieved in human tissue culture cells in a timeframe that is screening with single-cell RNA-seq, but this does not easily scale compatible with image-based phenotyping. We developed a pipe- to the profiling of thousands of single cells from thousands of line to construct a large-scale arrayed library of 2,281 sequence- perturbations, and is limited to features that can be read from verified CRISPR-Cas9 targeting plasmids and profiled this library RNA transcript profiles (Adamson et al, 2016; Dixit et al, 2016; for genes affecting cellular morphology and the subcellular local- Jaitin et al, 2016; Datlinger et al, 2017). Moreover, sequencing- ization of components of the nuclear pore complex (NPC).
    [Show full text]
  • A Minimal Set of Internal Control Genes for Gene Expression Studies in Head
    bioRxiv preprint doi: https://doi.org/10.1101/108381; this version posted February 14, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 A minimal set of internal control genes for gene expression studies in head 2 and neck squamous cell carcinoma. 1 1 1 2 3 Vinayak Palve , Manisha Pareek , Neeraja M Krishnan , Gangotri Siddappa , 2 2 1,3* 4 Amritha Suresh , Moni Abraham Kuriakose and Binay Panda 5 1 6 Ganit Labs, Bio-IT Centre, Institute of Bioinformatics and Applied Biotechnology, 7 Bangalore 560100 2 8 Mazumdar Shaw Cancer Centre and Mazumdar Shaw Centre for Translational 9 Research, Bangalore 560096 3 10 Strand Life Sciences, Bangalore 560024 11 * Corresponding author: [email protected] 12 13 Abstract: 14 Background: Selection of the right reference gene(s) is crucial in the analysis and 15 interpretation of gene expression data. In head and neck cancer, studies evaluating 16 the efficacy of internal reference genes are rare. Here, we present data for a minimal 17 set of candidates as internal control genes for gene expression studies in head and 18 neck cancer. 19 Methods: We analyzed data from multiple sources (in house whole-genome gene 20 expression microarrays, n=21; TCGA RNA-seq, n=42, and published gene 21 expression studies in head and neck tumors from literature) to come up with a set of 22 genes (discovery set) for their stable expression across tumor and normal tissues.
    [Show full text]
  • Mediator of DNA Damage Checkpoint 1 (MDC1) Is a Novel Estrogen Receptor Co-Regulator in Invasive 6 Lobular Carcinoma of the Breast 7 8 Evelyn K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.16.423142; this version posted December 16, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. 1 Running Title: MDC1 co-regulates ER in ILC 2 3 Research article 4 5 Mediator of DNA damage checkpoint 1 (MDC1) is a novel estrogen receptor co-regulator in invasive 6 lobular carcinoma of the breast 7 8 Evelyn K. Bordeaux1+, Joseph L. Sottnik1+, Sanjana Mehrotra1, Sarah E. Ferrara2, Andrew E. Goodspeed2,3, James 9 C. Costello2,3, Matthew J. Sikora1 10 11 +EKB and JLS contributed equally to this project. 12 13 Affiliations 14 1Dept. of Pathology, University of Colorado Anschutz Medical Campus 15 2Biostatistics and Bioinformatics Shared Resource, University of Colorado Comprehensive Cancer Center 16 3Dept. of Pharmacology, University of Colorado Anschutz Medical Campus 17 18 Corresponding author 19 Matthew J. Sikora, PhD.; Mail Stop 8104, Research Complex 1 South, Room 5117, 12801 E. 17th Ave.; Aurora, 20 CO 80045. Tel: (303)724-4301; Fax: (303)724-3712; email: [email protected]. Twitter: 21 @mjsikora 22 23 Authors' contributions 24 MJS conceived of the project. MJS, EKB, and JLS designed and performed experiments. JLS developed models 25 for the project. EKB, JLS, SM, and AEG contributed to data analysis and interpretation. SEF, AEG, and JCC 26 developed and performed informatics analyses. MJS wrote the draft manuscript; all authors read and revised the 27 manuscript and have read and approved of this version of the manuscript.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Deubiquitinases in Cancer: New Functions and Therapeutic Options
    Oncogene (2012) 31, 2373–2388 & 2012 Macmillan Publishers Limited All rights reserved 0950-9232/12 www.nature.com/onc REVIEW Deubiquitinases in cancer: new functions and therapeutic options JM Fraile1, V Quesada1, D Rodrı´guez, JMP Freije and C Lo´pez-Otı´n Departamento de Bioquı´mica y Biologı´a Molecular, Facultad de Medicina, Instituto Universitario de Oncologı´a, Universidad de Oviedo, Oviedo, Spain Deubiquitinases (DUBs) have fundamental roles in the Hunter, 2010). Consistent with the functional relevance ubiquitin system through their ability to specifically of proteases in these processes, alterations in their deconjugate ubiquitin from targeted proteins. The human structure or in the mechanisms controlling their genome encodes at least 98 DUBs, which can be grouped spatiotemporal expression patterns and activities cause into 6 families, reflecting the need for specificity in diverse pathologies such as arthritis, neurodegenerative their function. The activity of these enzymes affects the alterations, cardiovascular diseases and cancer. Accord- turnover rate, activation, recycling and localization ingly, many proteases are an important focus of of multiple proteins, which in turn is essential for attention for the pharmaceutical industry either as drug cell homeostasis, protein stability and a wide range of targets or as diagnostic and prognostic biomarkers signaling pathways. Consistent with this, altered DUB (Turk, 2006; Drag and Salvesen, 2010). function has been related to several diseases, including The recent availability of the genome sequence cancer. Thus, multiple DUBs have been classified as of different organisms has facilitated the identification oncogenes or tumor suppressors because of their regula- of their entire protease repertoire, which has been tory functions on the activity of other proteins involved in defined as degradome (Lopez-Otin and Overall, 2002).
    [Show full text]
  • Selective Androgen Receptor Modulators (Sarms) And
    (19) TZZ ¥6B_T (11) EP 2 222 636 B1 (12) EUROPEAN PATENT SPECIFICATION (45) Date of publication and mention (51) Int Cl.: of the grant of the patent: C07D 207/08 (2006.01) C07D 207/09 (2006.01) 10.04.2013 Bulletin 2013/15 C07D 498/04 (2006.01) A61K 31/402 (2006.01) A61P 5/26 (2006.01) (21) Application number: 08865188.0 (86) International application number: (22) Date of filing: 12.12.2008 PCT/US2008/013657 (87) International publication number: WO 2009/082437 (02.07.2009 Gazette 2009/27) (54) SELECTIVE ANDROGEN RECEPTOR MODULATORS (SARMS) AND USES THEREOF SELEKTIVE ANDROGENREZEPTORMODULATOREN (SARMS) UND IHRE VERWENDUNG MODULATEURS SÉLECTIFS DU RÉCEPTEUR ANDROGÈNE (SARM) ET LEURS UTILISATIONS (84) Designated Contracting States: (74) Representative: Baldock, Sharon Claire AT BE BG CH CY CZ DE DK EE ES FI FR GB GR Boult Wade Tennant HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT Verulam Gardens RO SE SI SK TR 70 Gray’s Inn Road Designated Extension States: London WC1X 8BT (GB) AL BA MK RS (56) References cited: (30) Priority: 21.12.2007 US 8731 P WO-A-02/068427 WO-A-03/090672 WO-A-2005/090282 WO-A-2006/124447 (43) Date of publication of application: WO-A-2007/015567 01.09.2010 Bulletin 2010/35 • HIGUCHI ET AL: "Potent, nonsteroidal selective (60) Divisional application: androgen receptor modulators (SARMs) based 12168231.4 / 2 489 656 on 8H-[1,4]oxazino[2,3-f]quinolin-8-ones" BIOORGANIC & MEDICINAL CHEMISTRY (73) Proprietor: Ligand Pharmaceuticals Inc.
    [Show full text]
  • Proteasome System of Protein Degradation and Processing
    ISSN 0006-2979, Biochemistry (Moscow), 2009, Vol. 74, No. 13, pp. 1411-1442. © Pleiades Publishing, Ltd., 2009. Original Russian Text © A. V. Sorokin, E. R. Kim, L. P. Ovchinnikov, 2009, published in Uspekhi Biologicheskoi Khimii, 2009, Vol. 49, pp. 3-76. REVIEW Proteasome System of Protein Degradation and Processing A. V. Sorokin*, E. R. Kim, and L. P. Ovchinnikov Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia; E-mail: [email protected]; [email protected] Received February 5, 2009 Abstract—In eukaryotic cells, degradation of most intracellular proteins is realized by proteasomes. The substrates for pro- teolysis are selected by the fact that the gate to the proteolytic chamber of the proteasome is usually closed, and only pro- teins carrying a special “label” can get into it. A polyubiquitin chain plays the role of the “label”: degradation affects pro- teins conjugated with a ubiquitin (Ub) chain that consists at minimum of four molecules. Upon entering the proteasome channel, the polypeptide chain of the protein unfolds and stretches along it, being hydrolyzed to short peptides. Ubiquitin per se does not get into the proteasome, but, after destruction of the “labeled” molecule, it is released and labels another molecule. This process has been named “Ub-dependent protein degradation”. In this review we systematize current data on the Ub–proteasome system, describe in detail proteasome structure, the ubiquitination system, and the classical ATP/Ub- dependent mechanism of protein degradation, as well as try to focus readers’ attention on the existence of alternative mech- anisms of proteasomal degradation and processing of proteins.
    [Show full text]
  • Genomic Amplification of Chromosome 20Q13.33 Is the Early Biomarker For
    Bui et al. BMC Medical Genomics 2020, 13(Suppl 10):149 https://doi.org/10.1186/s12920-020-00776-z RESEARCH Open Access Genomic amplification of chromosome 20q13.33 is the early biomarker for the development of sporadic colorectal carcinoma Vo-Minh-Hoang Bui1,2, Clément Mettling3, Jonathan Jou4 and H. Sunny Sun1,5* From The 18th Asia Pacific Bioinformatics Conference Seoul, Korea. 18-20 August 2020 Abstract Background: Colorectal carcinoma (CRC) is the third most common cancer in the world and also the third leading cause of cancer-related mortality in Taiwan. CRC tumorigenesis is a multistep process, starting from mutations causing loss of function of tumor suppressor genes, canonically demonstrated in adenomatous polyposis coli pathogenesis. Although many genes or chromosomal alterations have been shown to be involved in this process, there are still unrecognized molecular events within CRC tumorigenesis. Elucidating these mechanisms may help improve the management and treatment. Methods: In this study, we aimed to identify copy number alteration of the smallest chromosomal regions that is significantly associated with sporadic CRC tumorigenesis using high-resolution array-based Comparative Genomic Hybridization (aCGH) and quantitative Polymerase chain reaction (qPCR). In addition, microsatellite instability assay and sequencing-based mutation assay were performed to illustrate the initiation event of CRC tumorigenesis. Results: A total of 571 CRC patients were recruited and 377 paired CRC tissues from sporadic CRC cases were used to define the smallest regions with chromosome copy number changes. In addition, 198 colorectal polyps from 160 patients were also used to study the role of 20q13.33 gain in CRC tumorigenesis.
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • PSMD14 Is Over-Expressed in Human Endometrial Cancer
    Over-expression of proteasome 26S subunit, non-ATPase 14 in human endometrial cancer. Shahan Mamoor, MS1 [email protected] East Islip, NY USA Gynecologic cancers including cancers of the endometrium are a clinical problem1-4. We mined published microarray data5,6 to discover genes associated with endometrial cancers by comparing transcriptomes of the normal and hyperplastic endometrium to endometrial tumors from humans. We identified proteasome 26S subunit, non-ATPase 14, encoded by PSMD14, as among the most differentially expressed genes, transcriptome-wide, in cancers of the endometrium. PSMD14 was expressed at significantly higher levels in endometrial tumor tissues as compared to the endometrium. Importantly, in human endometrial cancer, primary tumor expression of PSMD14 was correlated with overall survival in white patients with low mutational burden. PSMD14 may be a molecule of interest in understanding the etiology or progression of human endometrial cancer. Keywords: endometrial cancer, gynecologic cancers, endometrium, PSMD14, proteasome 26S subunit, non-ATPase 14, systems biology of endometrial cancer, targeted therapeutics in endometrial cancer. 1 Endometrial cancer is the most common gynecologic cancer in the developed world1. Over the last three decades, the incidence of endometrial cancer has increased 21%4 and the death rate has increased 100%3. We harnessed the power of independently published microarray datasets5,6 to determine in an unbiased fashion and at the systems-level genes most differentially expressed in endometrial tumors. We report here the differential and increased expression of the proteasome 26S subunit, non-ATPase 14 (PSMD14) in human endometrial cancer. Methods We utilized datasets GSE636785 and GSE1061916 for this global differential gene expression analysis of human endometrial cancer in conjunction with GEO2R.
    [Show full text]