Molecular Subtypes of Diffuse Large B-Cell Lymphoma Arise by Distinct Genetic Pathways

Total Page:16

File Type:pdf, Size:1020Kb

Molecular Subtypes of Diffuse Large B-Cell Lymphoma Arise by Distinct Genetic Pathways Molecular subtypes of diffuse large B-cell lymphoma arise by distinct genetic pathways Georg Lenza, George W. Wrightb, N. C. Tolga Emrea, Holger Kohlhammera, Sandeep S. Davea, R. Eric Davisa, Shannon Cartya, Lloyd T. Lama, A. L. Shaffera, Wenming Xiaoc, John Powellc, Andreas Rosenwaldd, German Ottd,e, Hans Konrad Muller-Hermelinkd, Randy D. Gascoynef, Joseph M. Connorsf, Elias Campog, Elaine S. Jaffeh, Jan Delabiei, Erlend B. Smelandj,k, Lisa M. Rimszal, Richard I. Fisherm,n, Dennis D. Weisenburgero, Wing C. Chano, and Louis M. Staudta,q aMetabolism Branch, bBiometric Research Branch, cCenter for Information Technology, and hLaboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892; dDepartment of Pathology, University of Wu¨rzburg, 97080 Wu¨rzburg, Germany; eDepartment of Clinical Pathology, Robert-Bosch-Krankenhaus, 70376 Stuttgart, Germany; fBritish Columbia Cancer Agency, Vancouver, British Columbia, Canada V5Z 4E6; gHospital Clinic, University of Barcelona, 08036 Barcelona, Spain; iPathology Clinic and jInstitute for Cancer Research, Rikshospitalet Hospital, Oslo, Norway; kCentre for Cancer Biomedicine, Faculty Division the Norwegian Radium Hospital, N-0310, Oslo, Norway; lDepartment of Pathology, University of Arizona, Tucson, AZ 85724; mSouthwest Oncology Group, 24 Frank Lloyd Wright Drive, Ann Arbor, MI 48106 ; nJames P. Wilmot Cancer Center, University of Rochester, Rochester, NY 14642; and oDepartments of Pathology and Microbiology, University of Nebraska, Omaha, NE 68198 Edited by Ira Pastan, National Cancer Institute, National Institutes of Health, Bethesda, MD, and approved July 7, 2008 (received for review May 2, 2008) Gene-expression profiling has been used to define 3 molecular Recent studies using fluorescence in situ hybridization, con- subtypes of diffuse large B-cell lymphoma (DLBCL), termed germi- ventional comparative genomic hybridization (CGH), or low- nal center B-cell-like (GCB) DLBCL, activated B-cell-like (ABC) DLBCL, resolution array-based CGH (aCGH) have shown that certain and primary mediastinal B-cell lymphoma (PMBL). To investigate genetic aberrations occur at different frequencies among DLBCL whether these DLBCL subtypes arise by distinct pathogenetic subtypes (4, 9–11). The t(14, 18) translocation involving BCL2 and mechanisms, we analyzed 203 DLBCL biopsy samples by high- amplification of REL was detected in 45% and 16% of GCB resolution, genome-wide copy number analysis coupled with DLBCLs, respectively, but never in ABC DLBCLs (4). Roughly gene-expression profiling. Of 272 recurrent chromosomal aberra- one-quarter of ABC DLBCLs had trisomy 3 or gain/amplification tions that were associated with gene-expression alterations, 30 of chromosome arm 3q, but these abnormalities never were de- were used differentially by the DLBCL subtypes (P < 0.006). An tected in GCB DLBCLs (9, 10). ABC DLBCLs were characterized amplicon on chromosome 19 was detected in 26% of ABC DLBCLs further by frequent gain of 18q and loss of 6q (9, 10). but in only 3% of GCB DLBCLs and PMBLs. A highly up-regulated These relatively low-resolution techniques were unable to gene in this amplicon was SPIB, which encodes an ETS family pinpoint the presumed cancer-relevant target gene in most transcription factor. Knockdown of SPIB by RNA interference was instances. To overcome these limitations, we combined aCGH toxic to ABC DLBCL cell lines but not to GCB DLBCL, PMBL, or using high-density whole-genome microarrays with gene expres- myeloma cell lines, strongly implicating SPIB as an oncogene sion profiling. By this concerted approach, we identifiedchro- involved in the pathogenesis of ABC DLBCL. Deletion of the mosomal aberrations that were significantly more frequent in a INK4a/ARF tumor suppressor locus and trisomy 3 also occurred particular DLBCL subtype than in the others, and some of these aberrations were associated with clinical outcome. This analysis almost exclusively in ABC DLBCLs and was associated with inferior revealed oncogenic pathways that are used differentially by the outcome within this subtype. FOXP1 emerged as a potential DLBCL subtypes, reinforcing the view that they represent oncogene in ABC DLBCL that was up-regulated by trisomy 3 and by pathogenetically distinct diseases. more focal high-level amplifications. In GCB DLBCL, amplification of the oncogenic mir-17–92 microRNA cluster and deletion of the Results and Discussion tumor suppressor PTEN were recurrent, but these events did not Definition of Recurrently Altered Minimal Common Regions (MCRs). occur in ABC DLBCL. Together, these data provide genetic evidence aCGH was performed on 203 DLBCL samples, including 74 that the DLBCL subtypes are distinct diseases that use different ABC DLBCLs, 72 GCB DLBCLs, 31 PMBLs, and 26 unclassi- oncogenic pathways. fied DLBCLs from a previously studied cohort of patients (4). We used oligonucleotide microarrays covering the entire ge- gene-expression profiling ͉ oncogenes ͉ tumor suppressor genes ͉ nome at 5-kb average spacing, allowing us to delineate copy comparative genomic hybridization number changes with great precision. To identify chromosomal aberrations associated with gene-expression changes, we profiled iffuse large B-cell lymphoma (DLBCL) is the most common gene expression in the same samples and developed a statistical Dtype of non-Hodgkin’s lymphoma (1). DLBCL can be cured algorithm termed ‘‘Gene Expression and Dosage Integrator’’ using anthracycline-based chemotherapy regimens in only 40%– (GEDI) to merge the data sets (Fig. 1). For each case, GEDI 50% of patients, suggesting that DLBCL is a heterogeneous divides the aCGH data into intervals with statistically similar diagnostic category (2). Indeed, gene-expression profiling stud- gene dosage, and each interval is classified as an amplification, ies have distinguished 3 molecular subtypes of DLBCL known as ‘‘germinal center B-cell-like’’ (GCB) DLBCL, ‘‘activated B-cell- Author contributions: G.L. and L.M.S. designed research; G.L., N.C.T.E., H.K., S.S.D., R.E.D., like’’ (ABC) DLBCL, and ‘‘primary mediastinal B-cell lym- S.C., L.T.L., and A.L.S. performed research; A.R., G.O., H.K.M.-H., R.D.G., J.M.C., E.C., E.S.J., phoma’’ (PMBL) (3–7). GCB DLBCLs seem to arise from J.D., E.B.S., L.M.R., R.I.F., D.D.W., and W.C.C. contributed new reagents; G.L., G.W.W., W.X., normal germinal center B cells, whereas ABC DLBCLs may J.P., and L.M.S. analyzed data; and G.L., G.W.W., and L.M.S. wrote the paper. arise from postgerminal center B cells that are arrested during The authors declare no conflict of interest. plasmacytic differentiation, and PMBLs may arise from thymic This article is a PNAS Direct Submission. B cells (8). Patients with these DLBCL subtypes have signifi- qTo whom correspondence should be addressed. E-mail: [email protected] cantly different survival rates following chemotherapy (3–6), This article contains supporting information online at www.pnas.org/cgi/content/full/ leading to the current proposal that they represent distinct 0804295105/DCSupplemental. biological disease entities (8). © 2008 by The National Academy of Sciences of the USA 13520–13525 ͉ PNAS ͉ September 9, 2008 ͉ vol. 105 ͉ no. 36 www.pnas.org͞cgi͞doi͞10.1073͞pnas.0804295105 Downloaded by guest on September 26, 2021 Chromosomal position focus attention on more common abnormalities, we limited High resolution Amplification subsequent analysis to the 272 MCRs that occurred in 8 or more array-based Ͼ comparative Gain samples (i.e., 5% of ABC and GCB DLBCL cases) (Table S1). genomic Loss hybridization Double (aCGH) deletion Chromosomal Aberrations Associated with the DLBCL Subtype Dis- Identify tinction. To elucidate oncogenic mechanisms in the DLBCL Gene expression Minimal Common Regions profiling (MCRs) subtypes, we focused our analysis on MCRs that were differen- Correlate gene expression Associate MCRs tially represented among ABC DLBCLs, GCB DLBCLs, and with MCRs with the PMBLs. We further reasoned that such MCRs might be less DLBCL subtype distinction likely to be common copy number polymorphisms present in the (false discovery rate human population (14). { < 5%) MCR Based on a false discovery rate (FDR) of 0.05, we observed 30 MCRs associated with the subtype distinction, far in excess of Fig. 1. Schematic of the Gene Expression and Dosage Integration algorithm chance (Fig. S2 and Table S2). Some of these MCRs probably (see methods). represent similar biology, such as single and double loss of the INK4A/ARF locus, both of which were much more frequent in single-copy gain, single-copy deletion, homozygous deletion, or ABC DLBCLs (Fig. 2). is declared wild type [supporting information (SI) Fig. S1]. Next, Aberrations most characteristic of ABC DLBCL included GEDI defines MCRs of abnormal gene dosage by assembling trisomy 3, deletion of chromosome arm 6q, gain/amplification of segments that affect the same chromosomal region in different a region on chromosome arm 18q, deletion of the INK4a/ARF cases and that belong to the same dosage category (12, 13). tumor suppressor locus on chromosome 9, and gain/amplifica- Finally, the GEDI algorithm determines the degree to which tion of a 9-Mb region on chromosome 19. Aberrations prefer- each gene within an MCR is altered in expression and computes entially used by GCB DLBCLs included amplification of the a statistic, summarizing these correlations across all genes in mir-17–92 microRNA in the MIHG1 locus cluster on chromo- the MCR. some 13, gain/amplification of a 7.6-Mb region on chromosome The GEDI algorithm yielded a total of 1940 MCRs within the 12, deletion of the PTEN tumor suppressor gene on chromosome DLBCL data set, ranging in size from 5 kb, affecting a single 10, and amplification of the REL locus on chromosome 2. The gene, to more than 240 Mb, affecting a whole chromosome. Of most frequent chromosomal lesions in PMBL included amplifi- these MCRs, 719 showed a significant association with gene- cation of a telomeric region of chromosome 9p, monosomy 10, expression alterations, whereas 1221 did not.
Recommended publications
  • Cyclin D1/Cyclin-Dependent Kinase 4 Interacts with Filamin a and Affects the Migration and Invasion Potential of Breast Cancer Cells
    Published OnlineFirst February 28, 2010; DOI: 10.1158/0008-5472.CAN-08-1108 Tumor and Stem Cell Biology Cancer Research Cyclin D1/Cyclin-Dependent Kinase 4 Interacts with Filamin A and Affects the Migration and Invasion Potential of Breast Cancer Cells Zhijiu Zhong, Wen-Shuz Yeow, Chunhua Zou, Richard Wassell, Chenguang Wang, Richard G. Pestell, Judy N. Quong, and Andrew A. Quong Abstract Cyclin D1 belongs to a family of proteins that regulate progression through the G1-S phase of the cell cycle by binding to cyclin-dependent kinase (cdk)-4 to phosphorylate the retinoblastoma protein and release E2F transcription factors for progression through cell cycle. Several cancers, including breast, colon, and prostate, overexpress the cyclin D1 gene. However, the correlation of cyclin D1 overexpression with E2F target gene regulation or of cdk-dependent cyclin D1 activity with tumor development has not been identified. This suggests that the role of cyclin D1 in oncogenesis may be independent of its function as a cell cycle regulator. One such function is the role of cyclin D1 in cell adhesion and motility. Filamin A (FLNa), a member of the actin-binding filamin protein family, regulates signaling events involved in cell motility and invasion. FLNa has also been associated with a variety of cancers including lung cancer, prostate cancer, melanoma, human bladder cancer, and neuroblastoma. We hypothesized that elevated cyclin D1 facilitates motility in the invasive MDA-MB-231 breast cancer cell line. We show that MDA-MB-231 motility is affected by disturbing cyclin D1 levels or cyclin D1-cdk4/6 kinase activity.
    [Show full text]
  • Whole-Genome Microarray Detects Deletions and Loss of Heterozygosity of Chromosome 3 Occurring Exclusively in Metastasizing Uveal Melanoma
    Anatomy and Pathology Whole-Genome Microarray Detects Deletions and Loss of Heterozygosity of Chromosome 3 Occurring Exclusively in Metastasizing Uveal Melanoma Sarah L. Lake,1 Sarah E. Coupland,1 Azzam F. G. Taktak,2 and Bertil E. Damato3 PURPOSE. To detect deletions and loss of heterozygosity of disease is fatal in 92% of patients within 2 years of diagnosis. chromosome 3 in a rare subset of fatal, disomy 3 uveal mela- Clinical and histopathologic risk factors for UM metastasis noma (UM), undetectable by fluorescence in situ hybridization include large basal tumor diameter (LBD), ciliary body involve- (FISH). ment, epithelioid cytomorphology, extracellular matrix peri- ϩ ETHODS odic acid-Schiff-positive (PAS ) loops, and high mitotic M . Multiplex ligation-dependent probe amplification 3,4 5 (MLPA) with the P027 UM assay was performed on formalin- count. Prescher et al. showed that a nonrandom genetic fixed, paraffin-embedded (FFPE) whole tumor sections from 19 change, monosomy 3, correlates strongly with metastatic death, and the correlation has since been confirmed by several disomy 3 metastasizing UMs. Whole-genome microarray analy- 3,6–10 ses using a single-nucleotide polymorphism microarray (aSNP) groups. Consequently, fluorescence in situ hybridization were performed on frozen tissue samples from four fatal dis- (FISH) detection of chromosome 3 using a centromeric probe omy 3 metastasizing UMs and three disomy 3 tumors with Ͼ5 became routine practice for UM prognostication; however, 5% years’ metastasis-free survival. to 20% of disomy 3 UM patients unexpectedly develop metas- tases.11 Attempts have therefore been made to identify the RESULTS. Two metastasizing UMs that had been classified as minimal region(s) of deletion on chromosome 3.12–15 Despite disomy 3 by FISH analysis of a small tumor sample were found these studies, little progress has been made in defining the key on MLPA analysis to show monosomy 3.
    [Show full text]
  • Rabbit Anti-Human HIPPI Polyclonal Antibody (DPABH-04510) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Rabbit anti-Human HIPPI polyclonal antibody (DPABH-04510) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Antigen Description Required for the formation of cilia. Plays an indirect role in sonic hedgehog signaling, cilia being required for all activity of the hedgehog pathway (By similarity). Has pro-apoptotic function via its interaction with HIP1, leading to recruit caspase-8 (CASP8) and trigger apoptosis. Has the ability to bind DNA sequence motif 5-AAAGACATG-3 present in the promoter of caspase genes such as CASP1, CASP8 and CASP10, suggesting that it may act as a transcription regulator; however the relevance of such function remains unclear. Immunogen Recombinant fragment corresponding to a region within amino acids 71-361 of Human HIPPI (Uniprot ID: Q9NWB7). Isotype IgG Source/Host Rabbit Species Reactivity Human Purification Immunogen affinity purified Conjugate Unconjugated Applications WB, IHC-P Format Liquid Size 100 μl Buffer pH: 7.00; Constituents: 0.75% Glycine, 1.21% Tris, 10% Glycerol Preservative None Storage Store at -20°C or lower. Aliquot to avoid repeated freezing and thawing. GENE INFORMATION Gene Name IFT57 intraflagellar transport 58 homolog (Chlamydomonas) [ Homo sapiens ] Official Symbol IFT57 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 1 © Creative Diagnostics All Rights Reserved Synonyms IFT57; intraflagellar transport 57 homolog (Chlamydomonas); HIPPI; MHS4R2; ESRRBL1; intraflagellar transport
    [Show full text]
  • Early Growth Response 1 Regulates Hematopoietic Support and Proliferation in Human Primary Bone Marrow Stromal Cells
    Hematopoiesis SUPPLEMENTARY APPENDIX Early growth response 1 regulates hematopoietic support and proliferation in human primary bone marrow stromal cells Hongzhe Li, 1,2 Hooi-Ching Lim, 1,2 Dimitra Zacharaki, 1,2 Xiaojie Xian, 2,3 Keane J.G. Kenswil, 4 Sandro Bräunig, 1,2 Marc H.G.P. Raaijmakers, 4 Niels-Bjarne Woods, 2,3 Jenny Hansson, 1,2 and Stefan Scheding 1,2,5 1Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden; 2Lund Stem Cell Center, Depart - ment of Laboratory Medicine, Lund University, Lund, Sweden; 3Division of Molecular Medicine and Gene Therapy, Department of Labora - tory Medicine, Lund University, Lund, Sweden; 4Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands and 5Department of Hematology, Skåne University Hospital Lund, Skåne, Sweden ©2020 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol. 2019.216648 Received: January 14, 2019. Accepted: July 19, 2019. Pre-published: August 1, 2019. Correspondence: STEFAN SCHEDING - [email protected] Li et al.: Supplemental data 1. Supplemental Materials and Methods BM-MNC isolation Bone marrow mononuclear cells (BM-MNC) from BM aspiration samples were isolated by density gradient centrifugation (LSM 1077 Lymphocyte, PAA, Pasching, Austria) either with or without prior incubation with RosetteSep Human Mesenchymal Stem Cell Enrichment Cocktail (STEMCELL Technologies, Vancouver, Canada) for lineage depletion (CD3, CD14, CD19, CD38, CD66b, glycophorin A). BM-MNCs from fetal long bones and adult hip bones were isolated as reported previously 1 by gently crushing bones (femora, tibiae, fibulae, humeri, radii and ulna) in PBS+0.5% FCS subsequent passing of the cell suspension through a 40-µm filter.
    [Show full text]
  • TCTE1 Is a Conserved Component of the Dynein Regulatory Complex and Is Required for Motility and Metabolism in Mouse Spermatozoa
    TCTE1 is a conserved component of the dynein PNAS PLUS regulatory complex and is required for motility and metabolism in mouse spermatozoa Julio M. Castanedaa,b,1, Rong Huac,d,1, Haruhiko Miyatab, Asami Ojib,e, Yueshuai Guoc,d, Yiwei Chengc,d, Tao Zhouc,d, Xuejiang Guoc,d, Yiqiang Cuic,d, Bin Shenc, Zibin Wangc, Zhibin Huc,f, Zuomin Zhouc,d, Jiahao Shac,d, Renata Prunskaite-Hyyrylainena,g,h, Zhifeng Yua,i, Ramiro Ramirez-Solisj, Masahito Ikawab,e,k,2, Martin M. Matzuka,g,i,l,m,n,2, and Mingxi Liuc,d,2 aDepartment of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030; bResearch Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan; cState Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, People’s Republic of China; dDepartment of Histology and Embryology, Nanjing Medical University, Nanjing 210029, People’s Republic of China; eGraduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 5650871, Japan; fAnimal Core Facility of Nanjing Medical University, Nanjing 210029, People’s Republic of China; gCenter for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030; hFaculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu FI-90014, Finland; iCenter for Drug Discovery, Baylor College of Medicine, Houston, TX 77030; jWellcome Trust Sanger Institute, Hinxton CB10 1SA, United Kingdom; kThe Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 1088639, Japan; lDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030; mDepartment of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030; and nDepartment of Pharmacology, Baylor College of Medicine, Houston, TX 77030 Contributed by Martin M.
    [Show full text]
  • A Homozygous Nonsense Variant in IFT52 Is Associated with a Human Skeletal Ciliopathy
    Clin Genet 2016 © 2016 John Wiley & Sons A/S. Printed in Singapore. All rights reserved Published by John Wiley & Sons Ltd CLINICAL GENETICS doi: 10.1111/cge.12762 Short Report A homozygous nonsense variant in IFT52 is associated with a human skeletal ciliopathy a a Girisha K.M., Shukla A., Trujillano D., Bhavani G.S., Hebbar M., K. M. Girisha ,A.Shukla , Kadavigere R., Rolfs A. A homozygous nonsense variant in IFT52 is D. Trujillanob,G.S.Bhavania, associated with a human skeletal ciliopathy. M. Hebbara, R. Kadavigered Clin Genet 2016. © John Wiley & Sons A/S. Published by John Wiley & and A. Rolfsb,c Sons Ltd, 2016 aDepartment of Medical Genetics, Intraflagellar transport (IFT) is vital for the functioning of primary cilia. Kasturba Medical College, Manipal b Defects in several components of IFT complexes cause a spectrum of University, Manipal, India, Department of ciliopathies with variable involvement of skeleton, brain, eyes, ectoderm and Bioinformatics, Centogene AG, Rostock, Germany, cAlbrecht-Kossel-Institute for kidneys. We examined a child from a consanguineous family who had short Neuroregeneration, Medical University stature, narrow thorax, short hands and feet, postaxial polydactyly of hands, Rostock, Rostock, Germany, and pigmentary retinopathy, small teeth and skeletal dysplasia. The clinical dDepartment of Radiodiagnosis and phenotype of the child shows significant overlap with cranioectodermal Imaging, Kasturba Medical College, dysplasia type I (Sensenbrenner syndrome). Whole-exome sequencing Manipal University, Manipal, India revealed a homozygous nonsense variant p.R142* in IFT52 encoding an Key words: chondrodysplasia – IFT-B core complex protein as the probable cause of her condition. This is ciliopathy – exome sequencing – the first report of a human disease associated with IFT52.
    [Show full text]
  • SUPPLEMENTARY DATA Supplementary Table 1. Top Ten
    SUPPLEMENTARY DATA Supplementary Table 1. Top ten most highly expressed protein-coding genes in the EndoC-βH1 cell line. Expression levels provided for non-mitochondrial genes in EndoC-βH1 and the corresponding expression levels in sorted primary human β-cells (1). Ensembl gene ID Gene Name EndoC-βH1 [RPKM] Primary β cells [RPKM] ENSG00000254647.2 INS 8012.452 166347.111 ENSG00000087086.9 FTL 3090.7454 2066.464 ENSG00000100604.8 CHGA 2853.107 1113.162 ENSG00000099194.5 SCD 1411.631 238.714 ENSG00000118271.5 TTR 1312.8928 1488.996 ENSG00000184009.5 ACTG1 1108.0277 839.681 ENSG00000124172.5 ATP5E 863.42334 254.779 ENSG00000156508.13 EEF1A1 831.17316 637.281 ENSG00000112972.10 HMGCS1 719.7504 22.104 ENSG00000167552.9 TUBA1A 689.1415 511.699 ©2016 American Diabetes Association. Published online at http://diabetes.diabetesjournals.org/lookup/suppl/doi:10.2337/db16-0361/-/DC1 SUPPLEMENTARY DATA Supplementary Table 2. List of genes selected for inclusion in the primary screen. Expression levels in EndoC-βH1 and sorted primary human β-cells are shown for all genes targeted for silencing in the primary screen, ordered by locus association (1). For gene selection, the following criteria were applied: we first considered (1) all protein-coding genes within 1 Mb of a type 2 diabetes association signal that (2) had non-zero expression (RPKM > 0) in both EndoC-βH1 and primary human β-cells. Previous studies have shown cis-eQTLs to form a relatively tight, symmetrical distribution around the target-gene transcription start site, and a 1 Mb cut-off is thus likely to capture most effector transcripts subject to cis regulation (2-5).
    [Show full text]
  • SEC22A CRISPR/Cas9 KO Plasmid (M): Sc-435547
    SANTA CRUZ BIOTECHNOLOGY, INC. SEC22A CRISPR/Cas9 KO Plasmid (m): sc-435547 BACKGROUND APPLICATIONS The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and SEC22A CRISPR/Cas9 KO Plasmid (m) is recommended for the disruption of CRISPR-associated protein (Cas9) system is an adaptive immune response gene expression in mouse cells. defense mechanism used by archea and bacteria for the degradation of foreign genetic material (4,6). This mechanism can be repurposed for other 20 nt non-coding RNA sequence: guides Cas9 functions, including genomic engineering for mammalian systems, such as to a specific target location in the genomic DNA gene knockout (KO) (1,2,3,5). CRISPR/Cas9 KO Plasmid products enable the U6 promoter: drives gRNA scaffold: helps Cas9 identification and cleavage of specific genes by utilizing guide RNA (gRNA) expression of gRNA bind to target DNA sequences derived from the Genome-scale CRISPR Knock-Out (GeCKO) v2 library developed in the Zhang Laboratory at the Broad Institute (3,5). Termination signal Green Fluorescent Protein: to visually REFERENCES verify transfection CRISPR/Cas9 Knockout Plasmid CBh (chicken β-Actin 1. Cong, L., et al. 2013. Multiplex genome engineering using CRISPR/Cas hybrid) promoter: drives expression of Cas9 systems. Science 339: 819-823. 2A peptide: allows production of both Cas9 and GFP from the 2. Mali, P., et al. 2013. RNA-guided human genome engineering via Cas9. same CBh promoter Science 339: 823-826. Nuclear localization signal 3. Ran, F.A., et al. 2013. Genome engineering using the CRISPR-Cas9 system. Nuclear localization signal SpCas9 ribonuclease Nat. Protoc. 8: 2281-2308.
    [Show full text]
  • Mesenchymal Stem Cells from Multiple Myeloma Patients Display Distinct Genomic Profile As Compared with Those from Normal Donors
    Leukemia (2009) 23, 1515–1527 & 2009 Macmillan Publishers Limited All rights reserved 0887-6924/09 $32.00 www.nature.com/leu ORIGINAL ARTICLE Mesenchymal stem cells from multiple myeloma patients display distinct genomic profile as compared with those from normal donors M Garayoa1,5,6, JL Garcia1,2,6, C Santamaria3, A Garcia-Gomez1, JF Blanco4, A Pandiella1, JM Herna´ndez3, FM Sanchez-Guijo3,5, M-C del Can˜izo3,5, NC Gutie´rrez3, and JF San Miguel1,3,5 1Centro de Investigacio´n del Ca´ncer, Instituto de Biologı´a Molecular y Celular del Ca´ncer, Universidad de Salamanca-CSIC, Salamanca, Spain; 2Unidad de Investigacio´n, Instituto de Estudio de Ciencias de la Salud de Castilla y Leo´n (IECSCYL) – Hospital Universitario de Salamanca. Salamanca, Spain; 3Servicio de Hematologı´a. Hospital Universitario de Salamanca. Salamanca, Spain; 4Servicio de Traumatologı´a. Hospital Universitario de Salamanca. Salamanca, Spain and 5Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y Leo´n, Salamanca, Spain It is an open question whether in multiple myeloma (MM) bone directly by interactions of myeloma cells with BM stromal cells marrow stromal cells contain genomic alterations, which may and extracellular matrix proteins or indirectly by secretion of contribute to the pathogenesis of the disease. We conducted an array-based comparative genomic hybridization (array-CGH) soluble cytokine and growth factors by myeloma cells and/or analysis to compare the extent of unbalanced genomic altera- stromal cells. These interactions and growth factor circuits tions in mesenchymal stem cells from 21 myeloma patients ultimately lead to the activation of pleiotrophic signalling (MM-MSCs) and 12 normal donors (ND-MSCs) after in vitro cascades, which promote proliferation, cell survival, anti- culture expansion.
    [Show full text]
  • Cell Culture-Based Profiling Across Mammals Reveals DNA Repair And
    1 Cell culture-based profiling across mammals reveals 2 DNA repair and metabolism as determinants of 3 species longevity 4 5 Siming Ma1, Akhil Upneja1, Andrzej Galecki2,3, Yi-Miau Tsai2, Charles F. Burant4, Sasha 6 Raskind4, Quanwei Zhang5, Zhengdong D. Zhang5, Andrei Seluanov6, Vera Gorbunova6, 7 Clary B. Clish7, Richard A. Miller2, Vadim N. Gladyshev1* 8 9 1 Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard 10 Medical School, Boston, MA, 02115, USA 11 2 Department of Pathology and Geriatrics Center, University of Michigan Medical School, 12 Ann Arbor, MI 48109, USA 13 3 Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, 14 MI 48109, USA 15 4 Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 16 48109, USA 17 5 Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10128, USA 18 6 Department of Biology, University of Rochester, Rochester, NY 14627, USA 19 7 Broad Institute, Cambridge, MA 02142, US 20 21 * corresponding author: Vadim N. Gladyshev ([email protected]) 22 ABSTRACT 23 Mammalian lifespan differs by >100-fold, but the mechanisms associated with such 24 longevity differences are not understood. Here, we conducted a study on primary skin 25 fibroblasts isolated from 16 species of mammals and maintained under identical cell culture 26 conditions. We developed a pipeline for obtaining species-specific ortholog sequences, 27 profiled gene expression by RNA-seq and small molecules by metabolite profiling, and 28 identified genes and metabolites correlating with species longevity. Cells from longer-lived 29 species up-regulated genes involved in DNA repair and glucose metabolism, down-regulated 30 proteolysis and protein transport, and showed high levels of amino acids but low levels of 31 lysophosphatidylcholine and lysophosphatidylethanolamine.
    [Show full text]
  • COMPARATIVE GENOME-WIDE DNA METHYLATION ANALYSIS in MYOCARDIAL TISSUE from DONORS with and WITHOUT DOWN SYNDROME Romina B. Ceja
    bioRxiv preprint doi: https://doi.org/10.1101/2020.07.15.203075; this version posted July 15, 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-ND 4.0 International license. COMPARATIVE GENOME-WIDE DNA METHYLATION ANALYSIS IN MYOCARDIAL TISSUE FROM DONORS WITH AND WITHOUT DOWN SYNDROME Romina B. Cejasa, d, Jie Wangb, d, Rachael Hageman-Blairc, Song Liub and Javier G. Blancoa, e Authors’ affiliation: a Department of Pharmaceutical sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America b Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America c Department of Biostatistics, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, New York, United States of America d Both authors contributed equally to this work. e Corresponding author. [email protected]. Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 470 Pharmacy Building, Buffalo, NY 14214 – 8033, United States of America bioRxiv preprint doi: https://doi.org/10.1101/2020.07.15.203075; this version posted July 15, 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-ND 4.0 International license.
    [Show full text]
  • The Changing Chromatome As a Driver of Disease: a Panoramic View from Different Methodologies
    The changing chromatome as a driver of disease: A panoramic view from different methodologies Isabel Espejo1, Luciano Di Croce,1,2,3 and Sergi Aranda1 1. Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain 2. Universitat Pompeu Fabra (UPF), Barcelona, Spain 3. ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain *Corresponding authors: Luciano Di Croce ([email protected]) Sergi Aranda ([email protected]) 1 GRAPHICAL ABSTRACT Chromatin-bound proteins regulate gene expression, replicate and repair DNA, and transmit epigenetic information. Several human diseases are highly influenced by alterations in the chromatin- bound proteome. Thus, biochemical approaches for the systematic characterization of the chromatome could contribute to identifying new regulators of cellular functionality, including those that are relevant to human disorders. 2 SUMMARY Chromatin-bound proteins underlie several fundamental cellular functions, such as control of gene expression and the faithful transmission of genetic and epigenetic information. Components of the chromatin proteome (the “chromatome”) are essential in human life, and mutations in chromatin-bound proteins are frequently drivers of human diseases, such as cancer. Proteomic characterization of chromatin and de novo identification of chromatin interactors could thus reveal important and perhaps unexpected players implicated in human physiology and disease. Recently, intensive research efforts have focused on developing strategies to characterize the chromatome composition. In this review, we provide an overview of the dynamic composition of the chromatome, highlight the importance of its alterations as a driving force in human disease (and particularly in cancer), and discuss the different approaches to systematically characterize the chromatin-bound proteome in a global manner.
    [Show full text]