MUC1-C Activates the Nurd Complex to Drive Dedifferentiation of Triple

Total Page:16

File Type:pdf, Size:1020Kb

MUC1-C Activates the Nurd Complex to Drive Dedifferentiation of Triple Published OnlineFirst September 13, 2019; DOI: 10.1158/0008-5472.CAN-19-1034 Cancer Genome and Epigenome Research MUC1-C Activates the NuRD Complex to Drive Dedifferentiation of Triple-Negative Breast Cancer Cells Tsuyoshi Hata1, Hasan Rajabi1, Hidekazu Takahashi1, Yota Yasumizu1, Wei Li1, Caining Jin1, Mark D. Long2, Qiang Hu2, Song Liu2, Atsushi Fushimi1, Nami Yamashita1, Ling Kui1, Deli Hong1, Masaaki Yamamoto1, Masaaki Miyo1, Masayuki Hiraki1, Takahiro Maeda1, Yozo Suzuki1, Mehmet K. Samur1, and Donald Kufe1 Abstract The NuRD chromatin remodeling and deacetylation com- formed complexes with these NuRD components on the ESR1 plex, which includes MTA1, MBD3, CHD4, and HDAC1 promoter. Downregulating MUC1-C decreased MTA1/MBD3/ among other components, is of importance for development CHD4/HDAC1 occupancy and increased H3K27 acetylation and cancer progression. The oncogenic mucin 1 (MUC1) C- on the ESR1 promoter, with induction of ESR1 expression and terminal subunit (MUC1-C) protein activates EZH2 and BMI1 downstream estrogen response pathways. Targeting MUC1-C in the epigenetic reprogramming of triple-negative breast and these NuRD components also induced expression of cancer (TNBC). However, there is no known link between FOXA1, GATA3, and other markers associated with the lumi- MUC1-C and chromatin remodeling complexes. Here, we nal phenotype. These findings support a model in which showed that MUC1-C binds directly to the MYC HLH-LZ MUC1-C activates the NuRD complex to drive dedifferentia- domain and identified a previously unrecognized MUC1- tion and reprogramming of TNBC cells. C!MYC pathway that regulates the NuRD complex. MUC1-C/MYC complexes selectively activated the MTA1 and Significance: MUC1-C directly interacts with MYC to acti- MBD3 genes and posttranscriptionally induced CHD4 expres- vate the NuRD complex, mediating regulation of the estrogen sion in basal- but not luminal-type BC cells. In turn, MUC1-C receptor in triple-negative breast cancer cells. Introduction also activates the IKK!NF-kB p65 pathway, binds to NF-kB p65, and promotes expression of the downstream BCL-XL and ZEB1 The mucin 1 (MUC1) C-terminal subunit (MUC1-C) is an genes (8, 9). MUC1-C activates gene expression in part by increas- oncogenic transmembrane protein that is overexpressed in triple- ing TF occupancy on target genes and by recruiting the p300 negative breast carcinomas (TNBC; refs. 1, 2). MUC1-C interacts acetyltransferase (5, 6). Other studies have shown that MUC1-C with receptor tyrosine kinases, such as EGFR, at the cell membrane induces DNA methyltransferase 1 (DNMT1) and DNMT3b, and and transduces prosurvival signals to the nucleus (2). In this thereby DNA methylation and repression of the CDH1 tumor- capacity, MUC1-C directly interacts with transcription factors (TF) suppressor gene (TSG; refs. 3, 4). MUC1-C also induces expression and contributes to the regulation of their target genes (2–4). For of the BMI1, RING1, and RING2 components of the polycomb instance, MUC1-C associates with b-catenin/TCF4 and activates repressive complex 1 (PRC1) and contributes to BMI1-mediated the WNT pathway CCND1 and MYC target genes (5–7). MUC1-C repression of the CDKN2 TSG (3, 4). These observations were extended by the demonstration that MUC1-C induces the EZH2, 1Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. SUZ12, and EED subunits of PRC2 and binds directly to 2 Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive EZH2 (3, 4). In turn, MUC1-C increases H3K27 trimethylation Cancer Center, Buffalo, New York. on the BRCA1 TSG and downregulates BRCA1 expression (3, 4). In Note: Supplementary data for this article are available at Cancer Research concert with involvement in epigenetic repression of TSGs, MUC1- Online (http://cancerres.aacrjournals.org/). C has been linked to plasticity as characterized by induction of Current address for T. Hata, H. Takahashi, M. Yamamoto, M. Miyo, M. Hiraki, and the epithelial–mesenchymal transition (EMT), the cancer stem cell Y. Suzuki: Department of Gastroenterological Surgery, Graduate School of (CSC) state, and drug resistance (3, 4). These findings suggested Medicine, Osaka University, Suita, Osaka 565-0871, Japan; and current address that MUC1-C may play a role in other epigenetic pathways of gene for Y. Yasumizu and T. Maeda: Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan. repression and activation associated with hallmarks of the cancer cell. Of importance for the present work, there is no available Corresponding Author: Donald Kufe, Dana-Farber Cancer Institute, Harvard evidence for involvement of MUC1-C in the regulation of ATP- Medical School, 450 Brookline Avenue, D830, Boston, MA 02215. Phone: 617- 632-3141; Fax: 617-632-2934; E-mail: [email protected] dependent chromatin-remodeling complexes (10). The nucleosome remodeling and histone deacetylase (NuRD) Cancer Res 2019;79:5711–22 complex regulates chromatin assembly and reorganization, doi: 10.1158/0008-5472.CAN-19-1034 and is of importance for metazoan development and cancer Ó2019 American Association for Cancer Research. progression (10). The highly conserved NuRD complex consists www.aacrjournals.org 5711 Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst September 13, 2019; DOI: 10.1158/0008-5472.CAN-19-1034 Hata et al. of different subunits with enzymatic and nonenzymatic functions Hospital for Sick Children) of genomic DNA purified with the that broadly occupy active promoters and enhancers in different GenElute Mammalian Genomic DNA Miniprep Kit (Millipore cell types (11, 12). The core chromodomain-helicase-DNA- Sigma). Cells were monitored for Mycoplasma contamination binding protein 3 (CHD3) and CHD4 catalyze ATP-dependent using the MycoAlert Mycoplasma Detection Kit (Lonza). chromatin remodeling in association with activity of histone deacetylase 1 (HDAC1) and HDAC2 subunits. Nonenzymatic Tetracycline-inducible and stable gene silencing and NuRD components include, among others, the (i) methyl-CpG– expression binding domain 2 (MBD2) and MBD3, (ii) metastasis-associated MUC1shRNA (MISSION shRNA TRCN0000122938 and gene 1 (MTA1), MTA2, and MTA3, and (iii) retinoblastoma- shRNA#2 TRCN0000430218), MYCshRNA (MISSION shRNA binding protein 4 (RBBP4) and RBBP7 proteins. MBD3 links the TRCN0000039642 and shRNA#2 TRCN0000039640), MTA1shRNA histone deacetylase subcomplex containing MTA1, HDAC, and (MISSION shRNA TRCN0000230496), MBD3shRNA (MISSION RBBP proteins with the chromatin-remodeling subcomplex con- shRNA TRCN0000274441), CHD4shRNA (MISSION shRNA taining CHD4 and GATAD2A/B (11). Of these NuRD compo- TRCN0000021361), or a control scrambled shRNA (CshRNA, nents, MTA1 is widely overexpressed in human cancers by Millipore Sigma) was used in pLKO-puro vector or inserted into mechanisms associated, at least in part, with activation of the the pLKO-tet-puro vector (Plasmid #21915; Addgene). The TGFb1 and WNT/b-catenin pathways (13, 14). MTA1 acts as a MUC1-C cDNA was inserted into the pinducer20 vector (Plasmid corepressor by interacting with ERa and repressing ERa-activated #44012; Addgene). The viral vectors were produced in 293T cells gene transcription (15, 16). MTA1 associates with MBD3 and as described (9). Cells transduced with the vectors were selected recruits HDAC1 to deacetylate TFs, such as HIF1a, and regulates for growth in 1 to 3 mg/mL puromycin or 100 mg/mL geneticin. their transactivation functions (17). In addition, MTA1 interacts Cells were treated with 500 ng/mL doxycycline (DOX; Millipore with the CHD subunits in integrating their chromatin remodeling Sigma). functions (18). The pleotropic roles of MTA1 in regulating gene transcription extend to induction of EMT in cancer cells (13, 14) Quantitative reverse-transcription PCR and promotion of invasion and metastases (19). MTA1 has also Total cellular RNA was isolated using Trizol reagent (Invitro- been linked to chromatin remodeling during DNA repair by gen). cDNAs were synthesized using the High Capacity cDNA recruitment to sites of DNA lesions and interaction with Reverse Transcription Kit (Applied Biosystems). The cDNA sam- fi PARP1 (20). ples were ampli ed using the Power SYBR Green PCR Master Mix MUC1-C activates EZH2 and BMI1 in the epigenetic repro- (Applied Biosystems) and the CFX96 Real-Time PCR System gramming of TNBC cells; however, there is no known link (BIO-RAD) as described (9). Primers used for qRT-PCR are listed between MUC1-C and chromatin remodeling complexes, such in Supplementary Table S1. as NuRD. The present studies demonstrate that MUC1-C is Immunoblotting and immunoprecipitation necessary for induction of the MTA1, MBD3, and CHD4 subunits Total and nuclear lysates prepared from subconfluent cells were of the NuRD complex in TNBC cells. The results show that MUC1- subjected to immunoblot analysis using anti–MUC1-C (#HM- C interacts directly with MYC and thereby (i) activates the MTA1 1630-P1ABX; Thermo Fisher Scientific), anti-MYC (#ab32072; and MBD3 genes, and (ii) induces posttranscriptional expression Abcam), anti-ERa (#ab108398; Abcam), anti–b-actin (#A5441; of the CHD4 protein. Of functional significance, we demonstrate Sigma), anti-MTA1 (#5647), anti-MBD3 (#14540), anti-CHD4 that the MUC1-C!MYC!NuRD pathway drives luminal!basal (#11912), anti-HDAC1 (#5356), anti-SOX2 (#D6D9), anti-KLF4 dedifferentiation of TNBC cells. (#D1F2), anti-BMI1 (#D20B7), anti-CD44
Recommended publications
  • R&D Day for Investors and Analysts
    R&D Day for Investors and Analysts November 16, 2020 Seagen 2020 R&D Day Clay Siegall, Ph.D. Roger Dansey, M.D. Nancy Whiting, Pharm.D Megan O’Meara, M.D. Shyra Gardai, Ph.D. President & Chief Chief Medical Officer EVP, Corporate Strategy VP, Early Stage Executive Director, Executive Officer Alliances and Development Immunology Communications 2 Forward-Looking Statements Certain of the statements made in this presentation are forward looking, such as those, among others, relating to the Company’s potential to achieve the noted development and regulatory milestones in 2021 and in future periods; anticipated activities related to the Company’s planned and ongoing clinical trials; the potential for the Company’s clinical trials to support further development, regulatory submissions and potential marketing approvals in the U.S. and other countries; the opportunities for, and the therapeutic and commercial potential of ADCETRIS, PADCEV, TUKYSA, tisotumab vedotin and ladiratuzumab vedotin and the Company’s other product candidates and those of its licensees and collaborators; the potential to submit a BLA for accelerated approval of tisotumab vedotin; the potential for data from the EV-301 and EV-201 cohort 2 clinical trials to support additional regulatory approvals of PADCEV; the potential for the approval of TUKYSA by the EMA; the therapeutic potential of the Company’s SEA technology and of the Company’s early stage pipeline agents including SGN-B6A, SGN-STNV, SGN-CD228A, SEA-CD40, SEA-TGT, SEA-BCMA and SEA-CD70; as well as other statements that are not historical fact. Actual results or developments may differ materially from those projected or implied in these forward-looking statements.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • CAR- T Cell Immunotherapies
    CAR T Therapy Current Status Future Challenges Introduction Basics of CAR T Therapy Cytotoxic T Lymphocytes are Specific and Potent Effector Cells Ultrastructure of CTL-mediated apoptosis The CTL protrudes deeply into cytoplasm of melanoma cell 3 Peter Groscurth, and Luis Filgueira Physiology 1998;13:17-21 What is CAR T Therapy? • CAR T therapy is the name given to chimeric antigen receptor (CAR) genetically modified T cells that are designed to recognize specific antigens on tumor cells resulting in their activation and proliferation eventually resulting in significant and durable destruction of malignant cells • CAR T cells are considered “a living drug” since they tend to persist for long periods of time • CAR T cells are generally created from the patients own blood cells although this technology is evolving to develop “off the shelf” CAR T cells 4 CAR T cells: Mechanism of Action T cell Tumor cell CAR enables T cell to Expression of recognize tumor cell antigen CAR Viral DNA Insertion Antigen Tumor cell apoptosis CAR T cells multiply and release cytokines 5 Chimeric Antigen Receptors Antigen binding VH Antigen Binding Domain domain scFv Single-chain variable fragment (scFv) bypasses MHC antigen presentation, allowing direct activation of T cell by cancer cell antigens VL Hinge region Hinge region Essential for optimal antigen binding Costimulatory Domain: CD28 or 4-1BB Costimulatory Enhances proliferation, cytotoxicity and domain persistence of CAR T cells Activation Domains Signaling Domain: CD3-zeta chain CD3-zeta chain Proliferation
    [Show full text]
  • Novel Insights Into the Thaumarchaeota in the Deepest Oceans: Their Metabolism and Potential Adaptation Mechanisms
    Zhong et al. Microbiome (2020) 8:78 https://doi.org/10.1186/s40168-020-00849-2 RESEARCH Open Access Novel insights into the Thaumarchaeota in the deepest oceans: their metabolism and potential adaptation mechanisms Haohui Zhong1,2, Laura Lehtovirta-Morley3, Jiwen Liu1,2, Yanfen Zheng1, Heyu Lin1, Delei Song1, Jonathan D. Todd3, Jiwei Tian4 and Xiao-Hua Zhang1,2,5* Abstract Background: Marine Group I (MGI) Thaumarchaeota, which play key roles in the global biogeochemical cycling of nitrogen and carbon (ammonia oxidizers), thrive in the aphotic deep sea with massive populations. Recent studies have revealed that MGI Thaumarchaeota were present in the deepest part of oceans—the hadal zone (depth > 6000 m, consisting almost entirely of trenches), with the predominant phylotype being distinct from that in the “shallower” deep sea. However, little is known about the metabolism and distribution of these ammonia oxidizers in the hadal water. Results: In this study, metagenomic data were obtained from 0–10,500 m deep seawater samples from the Mariana Trench. The distribution patterns of Thaumarchaeota derived from metagenomics and 16S rRNA gene sequencing were in line with that reported in previous studies: abundance of Thaumarchaeota peaked in bathypelagic zone (depth 1000–4000 m) and the predominant clade shifted in the hadal zone. Several metagenome-assembled thaumarchaeotal genomes were recovered, including a near-complete one representing the dominant hadal phylotype of MGI. Using comparative genomics, we predict that unexpected genes involved in bioenergetics, including two distinct ATP synthase genes (predicted to be coupled with H+ and Na+ respectively), and genes horizontally transferred from other extremophiles, such as those encoding putative di-myo-inositol-phosphate (DIP) synthases, might significantly contribute to the success of this hadal clade under the extreme condition.
    [Show full text]
  • Galectin-3 Promotes Aβ Oligomerization and Aβ Toxicity in a Mouse Model of Alzheimer’S Disease
    Cell Death & Differentiation (2020) 27:192–209 https://doi.org/10.1038/s41418-019-0348-z ARTICLE Galectin-3 promotes Aβ oligomerization and Aβ toxicity in a mouse model of Alzheimer’s disease 1,2 1,3 1 1 1,2 4,5 Chih-Chieh Tao ● Kuang-Min Cheng ● Yun-Li Ma ● Wei-Lun Hsu ● Yan-Chu Chen ● Jong-Ling Fuh ● 4,6,7 3 1,2,3 Wei-Ju Lee ● Chih-Chang Chao ● Eminy H. Y. Lee Received: 1 October 2018 / Revised: 13 April 2019 / Accepted: 2 May 2019 / Published online: 24 May 2019 © ADMC Associazione Differenziamento e Morte Cellulare 2019. This article is published with open access Abstract Amyloid-β (Aβ) oligomers largely initiate the cascade underlying the pathology of Alzheimer’s disease (AD). Galectin-3 (Gal-3), which is a member of the galectin protein family, promotes inflammatory responses and enhances the homotypic aggregation of cancer cells. Here, we examined the role and action mechanism of Gal-3 in Aβ oligomerization and Aβ toxicities. Wild-type (WT) and Gal-3-knockout (KO) mice, APP/PS1;WT mice, APP/PS1;Gal-3+/− mice and brain tissues from normal subjects and AD patients were used. We found that Aβ oligomerization is reduced in Gal-3 KO mice injected with Aβ, whereas overexpression of Gal-3 enhances Aβ oligomerization in the hippocampi of Aβ-injected mice. Gal-3 expression shows an age-dependent increase that parallels endogenous Aβ oligomerization in APP/PS1 mice. Moreover, Aβ oligomerization, Iba1 expression, GFAP expression and amyloid plaque accumulation are reduced in APP/ PS1;Gal-3+/− mice compared with APP/PS1;WT mice.
    [Show full text]
  • MUC1-C Oncoprotein As a Target in Breast Cancer: Activation of Signaling Pathways and Therapeutic Approaches
    Oncogene (2013) 32, 1073–1081 & 2013 Macmillan Publishers Limited All rights reserved 0950-9232/13 www.nature.com/onc REVIEW MUC1-C oncoprotein as a target in breast cancer: activation of signaling pathways and therapeutic approaches DW Kufe Mucin 1 (MUC1) is a heterodimeric protein formed by two subunits that is aberrantly overexpressed in human breast cancer and other cancers. Historically, much of the early work on MUC1 focused on the shed mucin subunit. However, more recent studies have been directed at the transmembrane MUC1-C-terminal subunit (MUC1-C) that functions as an oncoprotein. MUC1-C interacts with EGFR (epidermal growth factor receptor), ErbB2 and other receptor tyrosine kinases at the cell membrane and contributes to activation of the PI3K-AKT and mitogen-activated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathways. MUC1-C also localizes to the nucleus where it activates the Wnt/b-catenin, signal transducer and activator of transcription (STAT) and NF (nuclear factor)-kB RelA pathways. These findings and the demonstration that MUC1-C is a druggable target have provided the experimental basis for designing agents that block MUC1-C function. Notably, inhibitors of the MUC1-C subunit have been developed that directly block its oncogenic function and induce death of breast cancer cells in vitro and in xenograft models. On the basis of these findings, a first-in-class MUC1-C inhibitor has entered phase I evaluation as a potential agent for the treatment of patients with breast cancers who express this oncoprotein. Oncogene (2013) 32, 1073–1081; doi:10.1038/onc.2012.158; published online 14 May 2012 Keywords: MUC1; breast cancer; oncoprotein; signaling pathways; targeted agents INTRODUCTION In breast tumor cells with loss of apical–basal polarity, the The mucin (MUC) family of high-molecular-weight glycoproteins MUC1-N/MUC1-C complex is found over the entire cell mem- 2 8 evolved in metazoans to provide protection for epithelial cell brane.
    [Show full text]
  • Membrane-Bound Mucins: the Mechanistic Basis for Alterations in the Growth and Survival of Cancer Cells
    Oncogene (2010) 29, 2893–2904 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc REVIEW Membrane-bound mucins: the mechanistic basis for alterations in the growth and survival of cancer cells S Bafna1, S Kaur1 and SK Batra1,2 1Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA and 2Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA Mucins (MUC) are high molecular weight O-linked tethered mucins are quite distinct from the classic glycoproteins whose primary functions are to hydrate, extracellular complex mucins forming the mucous layers protect, and lubricate the epithelial luminal surfaces of the of the gastrointestinal and respiratory tracts. These ducts within the human body. The MUC family is epithelial membrane-tethered mucins are the transmem- comprised of large secreted gel forming and transmem- brane (TM) molecules, expressed by most glandular and brane (TM) mucins. MUC1, MUC4, and MUC16 are the ductal epithelial cells (Taylor-Papadimitriou et al., well-characterized TM mucins and have been shown to be 1999). Several TM mucins (MUC1, MUC3A, MUC3B, aberrantly overexpressed in various malignancies includ- MUC4, MUC 12, MUC13, MUC15, MUC16, MUC17, ing cystic fibrosis, asthma, and cancer. Recent studies MUC20, and MUC21) (human mucins are designated have uncovered the unique roles of these mucins in the as MUC, whereas other species mucins are designated as pathogenesis of cancer. These mucins possess specific Muc) have been identified so far (Moniaux et al., 2001; domains that can make complex associations with various Chaturvedi et al., 2008a; Itoh et al., 2008).
    [Show full text]
  • (EMA) Is Preferentially Expressed by ALK Positive Anaplastic Large Cell Lymphoma, in The
    J Clin Pathol 2001;54:933–939 933 MUC1 (EMA) is preferentially expressed by ALK positive anaplastic large cell lymphoma, in the normally glycosylated or only partly J Clin Pathol: first published as on 1 December 2001. Downloaded from hypoglycosylated form R L ten Berge*, F G M Snijdewint*, S von MensdorV-Pouilly, RJJPoort-Keesom, J J Oudejans, J W R Meijer, R Willemze, J Hilgers, CJLMMeijer Abstract 30–50% of cases, a chromosomal aberration Aims—To investigate whether MUC1 such as the t(2;5)(p23;q35) translocation gives mucin, a high molecular weight trans- rise to expression of the anaplastic lymphoma membrane glycoprotein, also known as kinase (ALK) protein,2 identifying a subgroup epithelial membrane antigen (EMA), dif- of patients with systemic ALCL with excellent fers in its expression and degree of glyco- prognosis.3–6 ALK expression seems specific for sylation between anaplastic large cell systemic nodal ALCL; it is not found in classic lymphoma (ALCL) and classic Hodgkin’s Hodgkin’s disease (HD)7–11 or primary cutane- disease (HD), and whether MUC1 ous ALCL.7101213Morphologically, these lym- immunostaining can be used to diVerenti- phomas may closely resemble systemic ALCL, ate between CD30 positive large cell being also characterised by CD30 positive lymphomas. tumour cells with abundant cytoplasm, large Methods/Results—Using five diVerent irregular nuclei, and a prominent single monoclonal antibodies (E29/anti-EMA, nucleolus or multiple nucleoli.1 Clinically, DF3, 139H2, VU-4H5, and SM3) that however, classic HD and primary cutaneous distinguish between various MUC1 glyco- ALCL have a more favourable prognosis than 12 14–16 Department of forms, high MUC1 expression (50–95% of ALK negative systemic ALCL.
    [Show full text]
  • Epithelial Mucin-1 (MUCI) Expression and MA5 Anti-MUC1 Monoclonal Antibody Targeting in Multiple Myeloma I
    Vol. 5, 3065s-3072s, October 1999 (Suppl.) Clinical Cancer Research 3065s Epithelial Mucin-1 (MUCI) Expression and MA5 Anti-MUC1 Monoclonal Antibody Targeting in Multiple Myeloma I J. Burton, z D. Mishina, T. Cardillo, K. Lew, cGy/mCi of injected dose compared with 3099 cGy/mCi of A. Rubin, D. M. Goldenberg, and D. V. Gold tumor-absorbed dose delivered by nonspecific antibody. Garden State Cancer Center, Belleville, New Jersey 07109 [J. B., D. M., T. C., K. L., D. M. G., D. V. G.], and St. Joseph's Hospital and Introduction Medical Center, Paterson, New Jersey 07503 [A. R.] MM 3 is a B-cell malignancy that appears to result from the transformation and monoclonal expansion of a cell with char- acteristics of a plasma cell, i.e., a terminally differentiated B cell Abstract (1, 2). The expression by MM cells of certain non-B-cell anti- Multiple myeloma (MM) is the second most common gens also raises the possibility of the transformation of an hematological cancer in the United States. It is typically earlier, more multipotent lymphoid precursor cell. As with nor- incurable, even with myeloablative chemotherapy and stem- mal plasma cells, this degree of terminal differentiation is as- cell transplantation. The epithelial mucin-1 (MUC1) glyco- sociated with the complete or partial loss of certain B-cell- protein is expressed by normal and malignant epithelial cells associated antigens, such as surface immunoglobulin and CDs but has also been shown to be expressed by MM cells. MUC1 19-22 (CD19 is expressed on normal plasma cells; Ref. 3).
    [Show full text]
  • Characterization and Function of Medium and Large Extracellular
    bioRxiv preprint doi: https://doi.org/10.1101/623553; this version posted April 30, 2019. 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. Characterization and function of medium and large extracellular vesicles from plasma and urine by surface antigens and Annexin V Ko Igami1, 2, 3, Takeshi Uchiumi1*, Saori Ueda1, Kazuyuki Kamioka2, 4, Daiki Setoyama1, Kazuhito Gotoh1, Masaru Akimoto1, Shinya Matsumoto1 and Dongchon Kang1 1Department of Clinical Chemistry and Laboratory Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan. 2Kyushu Pro Search Limited Liability Partnership, 4-1, Kyudaishimmachi, Nishi- ku, Fukuoka, 819-0388, Japan. 3Business Management Division, Clinical Laboratory Business Segment, LSI Medience Corporation, 13-4, Uchikanda 1-chome, Chiyoda-ku, Tokyo, 101-8517, Japan. 4Medical Solutions Department, LSI Medience Corporation, 3- 30-1, Shimura, Itabashi-ku, Tokyo, 174-8555, Japan. Running title: Characterization of plasma and urine extracellular microvesicles Keywords: microvesicle, cell-derived microvesicle, peptidase, plasma, urine *Correspondence to: Takeshi Uchiumi, Department of Clinical Chemistry and Laboratory Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan. Tel: (+81) 92-642-5750; Fax: (+81) 92-642-5772 1 bioRxiv preprint doi: https://doi.org/10.1101/623553; this version posted April 30, 2019. 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.
    [Show full text]
  • Target Gene Gene Description Validation Diana Miranda
    Supplemental Table S1. Mmu-miR-183-5p in silico predicted targets. TARGET GENE GENE DESCRIPTION VALIDATION DIANA MIRANDA MIRBRIDGE PICTAR PITA RNA22 TARGETSCAN TOTAL_HIT AP3M1 adaptor-related protein complex 3, mu 1 subunit V V V V V V V 7 BTG1 B-cell translocation gene 1, anti-proliferative V V V V V V V 7 CLCN3 chloride channel, voltage-sensitive 3 V V V V V V V 7 CTDSPL CTD (carboxy-terminal domain, RNA polymerase II, polypeptide A) small phosphatase-like V V V V V V V 7 DUSP10 dual specificity phosphatase 10 V V V V V V V 7 MAP3K4 mitogen-activated protein kinase kinase kinase 4 V V V V V V V 7 PDCD4 programmed cell death 4 (neoplastic transformation inhibitor) V V V V V V V 7 PPP2R5C protein phosphatase 2, regulatory subunit B', gamma V V V V V V V 7 PTPN4 protein tyrosine phosphatase, non-receptor type 4 (megakaryocyte) V V V V V V V 7 EZR ezrin V V V V V V 6 FOXO1 forkhead box O1 V V V V V V 6 ANKRD13C ankyrin repeat domain 13C V V V V V V 6 ARHGAP6 Rho GTPase activating protein 6 V V V V V V 6 BACH2 BTB and CNC homology 1, basic leucine zipper transcription factor 2 V V V V V V 6 BNIP3L BCL2/adenovirus E1B 19kDa interacting protein 3-like V V V V V V 6 BRMS1L breast cancer metastasis-suppressor 1-like V V V V V V 6 CDK5R1 cyclin-dependent kinase 5, regulatory subunit 1 (p35) V V V V V V 6 CTDSP1 CTD (carboxy-terminal domain, RNA polymerase II, polypeptide A) small phosphatase 1 V V V V V V 6 DCX doublecortin V V V V V V 6 ENAH enabled homolog (Drosophila) V V V V V V 6 EPHA4 EPH receptor A4 V V V V V V 6 FOXP1 forkhead box P1 V
    [Show full text]
  • Human Urinary Exosomes As Innate Immune Effectors
    BASIC RESEARCH www.jasn.org Human Urinary Exosomes as Innate Immune Effectors † † ‡ Thomas F. Hiemstra,* Philip D. Charles, Tannia Gracia, Svenja S. Hester,§ † ‡ | Laurent Gatto, Rafia Al-Lamki,* R. Andres Floto,* Ya Su, Jeremy N. Skepper, † ‡ Kathryn S. Lilley, and Fiona E. Karet Frankl *Department of Medicine, †Cambridge Centre for Proteome Research and Cambridge Systems Biology Centre, Department of Biochemistry, ‡Department of Medical Genetics, and |Multi-Imaging Centre, Department of Anatomy, University of Cambridge, Cambridge, United Kingdom; and §Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom ABSTRACT Exosomes are small extracellular vesicles, approximately 50 nm in diameter, derived from the endocytic pathway and released by a variety of cell types. Recent data indicate a spectrum of exosomal functions, including RNA transfer, antigen presentation, modulation of apoptosis, and shedding of obsolete protein. Exosomes derived from all nephron segments are also present in human urine, where their function is unknown. Although one report suggested in vitro uptake of exosomes by renal cortical collecting duct cells, most studies of human urinary exosomes have focused on biomarker discovery rather than exosome function. Here, we report results from in-depth proteomic analyses and EM showing that normal human urinary exosomes are significantly enriched for innate immune proteins that include antimicrobial proteins and peptides and bacterial and viral receptors. Urinary exosomes, but not the prevalent soluble urinary protein uromodulin (Tamm–Horsfall protein), potently inhibited growth of pathogenic and commensal Escherichia coli and induced bacterial lysis. Bacterial killing depended on exosome structural integrity and occurred optimally at the acidic pH typical of urine from omnivorous humans.
    [Show full text]