PSMD5 Inactivation Promotes 26S Proteasome Assembly During Colorectal Tumor Progression Avi Levin1,2, Adi Minis1, Gadi Lalazar3, Jose Rodriguez1, and Hermann Steller1

Total Page:16

File Type:pdf, Size:1020Kb

PSMD5 Inactivation Promotes 26S Proteasome Assembly During Colorectal Tumor Progression Avi Levin1,2, Adi Minis1, Gadi Lalazar3, Jose Rodriguez1, and Hermann Steller1 Published OnlineFirst May 1, 2018; DOI: 10.1158/0008-5472.CAN-17-2296 Cancer Molecular Cell Biology Research PSMD5 Inactivation Promotes 26S Proteasome Assembly during Colorectal Tumor Progression Avi Levin1,2, Adi Minis1, Gadi Lalazar3, Jose Rodriguez1, and Hermann Steller1 Abstract Protein degradation by the ubiquitin–proteasome system (UPS) is central to protein homeostasis and cell survival. The active 26S proteasome is a PSMD5 large protease complex consist- Malignant ing of a catalytic 20S subunit transformation and 19S regulatory particles. PSMD5/S5b Cancer cells are exposed to con- down-regulation siderable protein overload due PSMD5 to high metabolic rates, repro- PSMD5 grammed energy metabolism, and aneuploidy. Here we report a mechanism that facilitates the Normal cell Malignant cell assembly of active 26S protea- somes in malignant cells. Upon Downregulation of PSMD5/S5b promotes 26S proteasome assembly, thereby increasing the tumorigenic transformation of proteolytic capacity of colorectal cancer cells, allowing them to maintain protein homeostasis. the gut epithelium, 26S pro- teasome assembly was signifi- © 2018 American Association for Cancer Research cantly enhanced, but levels of individual subunits were not changed. This enhanced assembly of 26S proteasomes increased further with tumor progression and was observed specifically in transformed cells, but not in other rapidly dividing cells. Moreover, expression of PSMD5, an inhibitor of proteasome assembly, was reduced in intestinal tumors and silenced with tumor progression. Reexpression of PSMD5 in tumor cells caused decreased 26S assembly and accumulation of polyubiquitinated proteins. These results suggest that inhibition of cancer-associated proteasome assembly may provide a novel therapeutic strategy to selectively kill cancer cells. Significance: Enhanced cancer-associated proteasome assembly is a major stress response that allows tumors to adapt to and to withstand protein overload. Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/13/3458/F1.large.jpg. Cancer Res; 78(13); 3458–68. Ó2018 AACR. Introduction vated protein synthesis can generate significant amounts of super- fluous and damaged proteins, which are potentially toxic. How- Protein synthesis and degradation are coordinated to allow ever, cells can increase their rates of protein degradation to adaptation to a rapidly changing microenvironment (1–3). Ele- maintain protein homeostasis (4, 5). Selective protein degrada- tion plays central roles in multiple biological processes including 1Strang Laboratory of Apoptosis and Cancer Biology, The Rockefeller University, removal of misfolded and potentially toxic proteins, control of New York, New York. 2Division of Gastroenterology, University of Iowa, Iowa cell-cycle progression, regulation of gene expression, and changes City, Iowa. 3Laboratory of Cellular Biophysics, The Rockefeller University, New in cell size and morphology (6, 7). The selective degradation of York, New York. most intracellular proteins is carried out by the ubiquitin-protea- Note: Supplementary data for this article are available at Cancer Research some system (UPS; refs. 8, 9). Proteins tagged with polyubiquitin Online (http://cancerres.aacrjournals.org/). chains are hydrolyzed into small peptides by the 26S proteasome Corresponding Authors: Hermann Steller, The Rockefeller University, 1230 York in an energy-dependent manner. The 26S proteasome is assem- Avenue, New York, NY 10065. Phone: 212-327-7075; Fax: 212-327-7076; E-mail: bled from a cylinder-shaped 20S proteolytic complex and a PA700 [email protected]; and Avi Levin, University of Iowa Hospitals and Clinics. (19S) ATPase-regulatory complex (10). The assembly and activity 200 Hawkins Drive, Iowa City, IA 52242. Phone: 319-356-7368; E-mail: of the 26S proteasome is tightly regulated by a large number of [email protected] cofactors and chaperones (8). Cancer cells are more exposed to doi: 10.1158/0008-5472.CAN-17-2296 proteotoxic stress due to high metabolic rates, reprogrammed Ó2018 American Association for Cancer Research. energy metabolism, and aneuploidy (5, 11–13). Despite this 3458 Cancer Res; 78(13) July 1, 2018 Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst May 1, 2018; DOI: 10.1158/0008-5472.CAN-17-2296 Enhanced Proteasome Assembly Mediates Tumor Proteostasis pressure, they successfully proliferate and efficiently withstand this several modifications: large bowels with and without tumors were challenge by fully exploiting the cells' defense mechanisms against flushed with cold PBS and opened longitudinally, washed with proteotoxic stress. For instance, constitutive activation of the heat 70% ethanol. Large-bowel tumors were dissected from normal shock factor protein 1 (HSF1)-regulated heat shock response large bowel. Normal large bowels and tumors were agitated in a (HSR) pathway is among the recurring features of malignant cells, solution of 2 mmol/L EDTA in PBS for 30 minutes at 4C. Tumors and inhibition of molecular chaperones represents a promising were dissociated using 70-mm strainer. therapeutic strategy for cancer treatment (14). In addition, Crypts were collected as described and embedded onto Matrigel increased proteasome activity can suppress aneuploidy-associated (BD Biosciences) with 50 mL/well in 24-well plates. DMEM/F12 proteotoxic stress (12). Activation of proteasomal degradation by culture medium (Gibco), containing Glutamax (Gibco) and pen- deletion of the proteasome-associated deubiquitylating enzyme icillin/streptomycin was supplemented with B27 (Gibco, 1:50), UBP6 attenuates the aneuploidy-induced changes in cellular pro- 100 ng/mL murine Noggin (Peprotech), 20 ng/mL murine EGF tein composition and improves cell fitness (15). The mechanisms (Peprotech), 10 ng/mL human basic-FGF (Peprotech) and 250 ng/ by which cancer cells overcome proteotoxic stress are of consid- mL human R-Spondin1 (Peprotech), 100 ng/mL mouse Wnt-3a erable scientific and clinical interest as advances in this area may (Peprotech) and 10 mmol/L nicotinamide (Sigma) for large-bowel identify new therapeutic targets. Notably, proteasome inhibitors organoids. Tumor organoids were cultured in medium containing have antitumor activity and are clinically used for the treatment of only B27 (Gibco, 1:50), 100 ng/mL murine Noggin (Peprotech) multiple myeloma and mantle cell lymphoma (16, 17). and 20 ng/mL murine EGF (Peprotech) supplements. The rate-limiting step in proteolysis by the UPS is considered to Lentiviral infection was used to transduce genes into advanced be substrate ubiquitination. However, recent findings indicate that tumor organoids. We generated recombinant lentiviral particles proteolysis rate is significantly changed by regulatory pathways expressing mouse PSMD5 in HEK293T. Because lentivirus parti- that affect the activity of the 26S proteasome itself, for example, by cles cannot penetrate the Matrigel layer, we incubated dissociated posttranslational modifications of proteasome subunits, or by organoids with viral particles in liquid media for 6 hours 37C facilitating the assembly of the 26S proteasome from the core under 5% CO2 (v/v). Next, cells were embedded in Matrigel and proteasome and the regulatory complexes (18, 19). It has been grown into organoids. Two to 3 days after plating of cells, previously shown that colon cancer cells increase expression of puromycin was added (5 mg/mL) to the medium for selection of proteasome subunits in a concerted manner (20). However, upre- successfully transduced cells. gulating the levels of proteasome subunits, while important, is not sufficient to significantly increase proteasome activity. Indeed, the Protein translation rate levels of functional 26S proteasomes depend not only on the The rate of protein translation was measured using the nonra- expression of its subunits, but also on their precise assembly. Work dioactive surface Sensing of translation (SUnSET) labeling method in yeast clearly demonstrates that assembly of 26S can vary (25). Briefly, puromycin (4 mmol/L) was added to culture medium significantly to meet the changing proteolytic needs of a cell (21). of organoids for 30 minutes, then organoids were harvested and Here we investigated a possible role of 26S proteasome assem- puromycin incorporation was detected by Western blot analysis bly for protein homeostasis during tumorigenesis of the mouse using an anti-puromycin antibody, clone 12D10 (1:5,000, Sigma). gut epithelium. We show that 26S proteasome assembly is highly To label new synthesized proteins in vivo, we injected into peri- increased upon tumorigenic transformation of the gut epitheli- toneum puromycin (0.04 mmol/gr of body weight of mice). We um. Importantly, this increase in 26S assembly is cell-autono- euthanized animals 30 minutes after puromycin injection and mous and occurs exclusively in transformed cells, but not in other analyzed tissues by Western blot analysis using an anti-puromycin rapidly dividing cells. In addition, levels of the proteasome antibody, clone 12D10 (1:5,000, Sigma). assembly inhibitor PSMD5/S5B are significantly decreased in tumors, and reexpression of PSMD5 blocks 26S proteasome Cell culture and DNA transfection assembly. Collectively, these results suggest that intestinal cancer HEK293T and HT-29 cells (purchased from ATCC) were grown cells escape proteotoxic stress by reducing PSMD5 to stimulate in DMEM containing 10% FBS and penicillin/streptomycin
Recommended publications
  • 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]
  • 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]
  • 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]
  • 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]
  • Produktinformation
    Produktinformation Diagnostik & molekulare Diagnostik Laborgeräte & Service Zellkultur & Verbrauchsmaterial Forschungsprodukte & Biochemikalien Weitere Information auf den folgenden Seiten! See the following pages for more information! Lieferung & Zahlungsart Lieferung: frei Haus Bestellung auf Rechnung SZABO-SCANDIC Lieferung: € 10,- HandelsgmbH & Co KG Erstbestellung Vorauskassa Quellenstraße 110, A-1100 Wien T. +43(0)1 489 3961-0 Zuschläge F. +43(0)1 489 3961-7 [email protected] • Mindermengenzuschlag www.szabo-scandic.com • Trockeneiszuschlag • Gefahrgutzuschlag linkedin.com/company/szaboscandic • Expressversand facebook.com/szaboscandic PSMD10 Antibody, Biotin conjugated Product Code CSB-PA018899LD01HU Abbreviation 26S proteasome non-ATPase regulatory subunit 10 Storage Upon receipt, store at -20°C or -80°C. Avoid repeated freeze. Uniprot No. O75832 Immunogen Recombinant Human 26S proteasome non-ATPase regulatory subunit 10 protein (1-226AA) Raised In Rabbit Species Reactivity Human Tested Applications ELISA Relevance Acts as a chaperone during the assembly of the 26S proteasome, specifically of the PA700/19S regulatory complex (RC). In the initial step of the base subcomplex assembly is part of an intermediate PSMD10:PSMC4:PSMC5:PAAF1 module which probably assembles with a PSMD5:PSMC2:PSMC1:PSMD2 module. Independently of the proteasome, regulates EGF-induced AKT activation through inhibition of the RHOA/ROCK/PTEN pahway, leading to prolonged AKT activation. Plays an important role in RAS-induced tumorigenesis. Acts as an proto-oncoprotein by being involved in negative regulation of tumor suppressors RB1 and p53/TP53. Overexpression is leading to phosphorylation of RB1 and proteasomal degradation of RB1. Regulates CDK4-mediated phosphorylation of RB1 by competing with CDKN2A for binding with CDK4. Facilitates binding of MDM2 to p53/TP53 and the mono- and polyubiquitination of p53/TP53 by MDM2 suggesting a function in targeting the TP53:MDM2 complex to the 26S proteasome.
    [Show full text]
  • The Proteasomal Deubiquitinating Enzyme PSMD14 Regulates 2 Macroautophagy by Controlling Golgi-To-ER Retrograde Transport
    bioRxiv preprint doi: https://doi.org/10.1101/2020.01.29.925503; this version posted January 31, 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-ND 4.0 International license. 1 The proteasomal deubiquitinating enzyme PSMD14 regulates 2 macroautophagy by controlling Golgi-to-ER retrograde transport 3 Bustamante HA1,2, Cereceda K3, González AE1,4, Valenzuela GE5,6, 4 Cheuquemilla Y6, Hernández S3, Arias-Muñoz E3, Cerda-Troncoso C3, 5 Bandau S8, Soza A3, Kausel G5, Kerr B3, Mardones GA1,7, Cancino J3, Hay 6 RT8, Rojas-Fernandez A6,8¥, Burgos PV3,9¥ 7 1Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, 5110566, Valdivia, Chile 8 2Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, 5110566, Valdivia, 9 Chile 10 3Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San 11 Sebastián, Lota 2465, 7510157, Santiago, Chile 12 4Institute of Biochemistry II, School of Medicine, Goethe University Frankfurt, Theoder-Stern-Kai 7, 60590, 13 Frankfurt am Main, Germany 14 5Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, 5110566, 15 Valdivia, Chile. 16 6Instituto de Medicina & Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad 17 Austral de Chile, 5110566, Valdivia, Chile. 18 7Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, 5110566, 19 Valdivia, Chile. 20 8Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, DD1 4HN, 21 Dundee, United Kingdom.
    [Show full text]
  • Cancer Vulnerabilities Unveiled by Genomic Loss
    Cancer Vulnerabilities Unveiled by Genomic Loss The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters. Citation Nijhawan, Deepak, Travis I. Zack, Yin Ren, Matthew R. Strickland, Rebecca Lamothe, Steven E. Schumacher, Aviad Tsherniak, et al. “Cancer Vulnerabilities Unveiled by Genomic Loss.” Cell 150, no. 4 (August 2012): 842–854. © 2012 Elsevier Inc. As Published http://dx.doi.org/10.1016/j.cell.2012.07.023 Publisher Elsevier B.V. Version Final published version Citable link http://hdl.handle.net/1721.1/91899 Terms of Use Article is made available in accordance with the publisher's policy and may be subject to US copyright law. Please refer to the publisher's site for terms of use. Cancer Vulnerabilities Unveiled by Genomic Loss Deepak Nijhawan,1,2,7,9,10 Travis I. Zack,1,2,3,9 Yin Ren,5 Matthew R. Strickland,1 Rebecca Lamothe,1 Steven E. Schumacher,1,2 Aviad Tsherniak,2 Henrike C. Besche,4 Joseph Rosenbluh,1,2,7 Shyemaa Shehata,1 Glenn S. Cowley,2 Barbara A. Weir,2 Alfred L. Goldberg,4 Jill P. Mesirov,2 David E. Root,2 Sangeeta N. Bhatia,2,5,6,7,8 Rameen Beroukhim,1,2,7,* and William C. Hahn1,2,7,* 1Departments of Cancer Biology and Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA 2Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA 3Biophysics Program, Harvard University, Boston, MA 02115, USA 4Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA 5Harvard-MIT Division of Health Sciences and Technology 6David H.
    [Show full text]
  • PSMD10 Antibody (Center) Purified Rabbit Polyclonal Antibody (Pab) Catalog # AW5126
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 PSMD10 Antibody (Center) Purified Rabbit Polyclonal Antibody (Pab) Catalog # AW5126 Specification PSMD10 Antibody (Center) - Product Information Application IF, WB, IHC-P,E Primary Accession O75832 Other Accession Q9Z2X2 Reactivity Human Predicted Mouse Host Rabbit Clonality Polyclonal Calculated MW H=24,16;M=25;Ra t=25 KDa Isotype Rabbit Ig Antigen Source HUMAN PSMD10 Antibody (Center) - Additional Information Gene ID 5716 Fluorescent image of Hela cells stained with PSMD10 Antibody (Center)(Cat#AW5126). Antigen Region AW5126 was diluted at 1:25 dilution. An 43-76 Alexa Fluor 488-conjugated goat anti-rabbit lgG at 1:400 dilution was used as the Other Names secondary antibody (green). Cytoplasmic 26S proteasome non-ATPase regulatory subunit 10, 26S proteasome regulatory actin was counterstained with Alexa Fluor® subunit p28, Gankyrin, p28(GANK), PSMD10 555 conjugated with Phalloidin (red). Dilution IF~~1:25 WB~~ 1:1000 IHC-P~~1:25 Target/Specificity This PSMD10 antibody is generated from a rabbit immunized with a KLH conjugated synthetic peptide between 43-76 amino acids from the Central region of human PSMD10. Format Purified polyclonal antibody supplied in PBS with 0.09% (W/V) sodium azide. This antibody is purified through a protein A column, followed by peptide affinity purification. Western blot analysis of lysates from MCF-7, PC-3, K562 cell line (from left to right), using Storage PSMD10 Antibody (Center)(Cat. #AW5126). Page 1/3 10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 Maintain refrigerated at 2-8°C for up to 2 AW5126 was diluted at 1:1000 at each lane.
    [Show full text]
  • Functional Gene Clusters in Global Pathogenesis of Clear Cell Carcinoma of the Ovary Discovered by Integrated Analysis of Transcriptomes
    International Journal of Environmental Research and Public Health Article Functional Gene Clusters in Global Pathogenesis of Clear Cell Carcinoma of the Ovary Discovered by Integrated Analysis of Transcriptomes Yueh-Han Hsu 1,2, Peng-Hui Wang 1,2,3,4,5 and Chia-Ming Chang 1,2,* 1 Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan; [email protected] (Y.-H.H.); [email protected] (P.-H.W.) 2 School of Medicine, National Yang-Ming University, Taipei 112, Taiwan 3 Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan 4 Department of Medical Research, China Medical University Hospital, Taichung 440, Taiwan 5 Female Cancer Foundation, Taipei 104, Taiwan * Correspondence: [email protected]; Tel.: +886-2-2875-7826; Fax: +886-2-5570-2788 Received: 27 April 2020; Accepted: 31 May 2020; Published: 2 June 2020 Abstract: Clear cell carcinoma of the ovary (ovarian clear cell carcinoma (OCCC)) is one epithelial ovarian carcinoma that is known to have a poor prognosis and a tendency for being refractory to treatment due to unclear pathogenesis. Published investigations of OCCC have mainly focused only on individual genes and lack of systematic integrated research to analyze the pathogenesis of OCCC in a genome-wide perspective. Thus, we conducted an integrated analysis using transcriptome datasets from a public domain database to determine genes that may be implicated in the pathogenesis involved in OCCC carcinogenesis. We used the data obtained from the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) DataSets. We found six interactive functional gene clusters in the pathogenesis network of OCCC, including ribosomal protein, eukaryotic translation initiation factors, lactate, prostaglandin, proteasome, and insulin-like growth factor.
    [Show full text]
  • Correction1 4784..4785
    Correction Correction: PCI-24781 Induces Caspase and Reactive Oxygen Species-Dependent Apoptosis In the article on PCI-24781 induces caspase and reactive oxygen species-dependent apoptosis published in the May 15, 2009 issue of Clinical Cancer Research, there was an error in Table 1. Down-regulated genes were incorrectly labeled as up-regulated genes. The correct table appears here. Bhalla S, Balasubramanian S, David K, et al. PCI-24781 induces caspase and reactive oxygen species-dependent apoptosis through NF-nB mechanisms and is synergistic with bortezomib in lymphoma cells. Clin Cancer Res 2009;15:3354–65. Table 1. Selected genes from expression analysis following 24-h treatment with PCI-24781, bortezomib, or the combination (in Ramos cells) Accn # Down-regulated genes 0.25 Mmol/L PCI/3 nmol/L Bor Name PCI-24781 Bortezomib Combination* Cell cycle-related NM_000075 Cyclin-dependent kinase 4 (CDK4) 0.49 0.83 0.37 NM_001237 Cyclin A2 (CCNA2) 0.43 0.87 0.37 NM_001950 E2F transcription factor 4, p107/p130-binding (E2F4) 0.48 0.79 0.40 NM_001951 E2F transcription factor 5, p130-binding (E2F5) 0.46 0.98 0.43 NM_003903 CDC16 cell division cycle 16 homolog (S cerevisiae) (CDC16) 0.61 0.78 0.43 NM_031966 Cyclin B1 (CCNB1) 0.55 0.90 0.43 NM_001760 Cyclin D3 (CCND3) 0.48 1.02 0.46 NM_001255 CDC20 cell division cycle 20 homolog (S cerevisiae; CDC20) 0.61 0.82 0.46 NM_001262 Cyclin-dependent kinase inhibitor 2C (p18, inhibits CDK4; CDKN2C) 0.61 1.15 0.56 NM_001238 Cyclin E1 (CCNE1) 0.56 1.05 0.60 NM_001239 Cyclin H (CCNH) 0.74 0.90 0.64 NM_004701
    [Show full text]
  • Comparative Transcriptomics Identifies Potential Stemness-Related Markers for Mesenchymal Stromal/Stem Cells
    bioRxiv preprint doi: https://doi.org/10.1101/2021.05.25.445659; this version posted May 26, 2021. 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-ND 4.0 International license. Comparative Transcriptomics Identifies Potential Stemness-Related Markers for Mesenchymal Stromal/Stem Cells Authors Myret Ghabriel 1, Ahmed El Hosseiny 1, 2, Ahmed Moustafa*1, 2 and Asma Amleh*1, 2 Affiliations 1Biotechnology Program, American University in Cairo, New Cairo 11835, Egypt 2Department of Biology, American University in Cairo, New Cairo 11835, Egypt *Corresponding authors: Ahmed Moustafa [email protected] Asma Amleh [email protected]. Abstract Mesenchymal stromal/stem cells (MSCs) are multipotent cells residing in multiple tissues with the capacity for self-renewal and differentiation into various cell types. These properties make them promising candidates for regenerative therapies. MSC identification is critical in yielding pure populations for successful therapeutic applications; however, the criteria for MSC identification proposed by the International Society for Cellular Therapy (ISCT) is inconsistent across different tissue sources. In this study, we aimed to identify potential markers to be used together with the ISCT’s criteria to provide a more accurate means of MSC identification. Thus, we carried out a comparative analysis of the expression of human and mouse MSCs derived from multiple tissues to identify the common differentially expressed genes. We show that six members of the proteasome degradation system are similarly expressed across MSCs derived from bone marrow, adipose tissue, amnion, and umbilical cord.
    [Show full text]
  • Genome-Wide Transcript and Protein Analysis Reveals Distinct Features of Aging in the Mouse Heart
    bioRxiv preprint doi: https://doi.org/10.1101/2020.08.28.272260; this version posted April 21, 2021. 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-ND 4.0 International license. Genome-wide transcript and protein analysis reveals distinct features of aging in the mouse heart Isabela Gerdes Gyuricza1, Joel M. Chick2, Gregory R. Keele1, Andrew G. Deighan1, Steven C. Munger1, Ron Korstanje1, Steven P. Gygi3, Gary A. Churchill1 1The Jackson Laboratory, Bar Harbor, Maine 04609 USA; 2Vividion Therapeutics, San Diego, California 92121, USA; 3Harvard Medical School, Boston, Massachusetts 02115, USA Corresponding author: [email protected] Key words for online indexing: Heart Aging Transcriptomics Proteomics eQTL pQTL Stoichiometry ABSTRACT Investigation of the molecular mechanisms of aging in the human heart is challenging due to confounding factors, such as diet and medications, as well limited access to tissues. The laboratory mouse provides an ideal model to study aging in healthy individuals in a controlled environment. However, previous mouse studies have examined only a narrow range of the genetic variation that shapes individual differences during aging. Here, we analyzed transcriptome and proteome data from hearts of genetically diverse mice at ages 6, 12 and 18 months to characterize molecular changes that occur in the aging heart. Transcripts and proteins reveal distinct biological processes that are altered through the course of natural aging. Transcriptome analysis reveals a scenario of cardiac hypertrophy, fibrosis, and reemergence of fetal gene expression patterns.
    [Show full text]