Identification of Novel Genes in BRCA1-Regulated Pathways

Total Page:16

File Type:pdf, Size:1020Kb

Identification of Novel Genes in BRCA1-Regulated Pathways Identification of Novel Genes in BRCA1-Regulated Pathways DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Shweta Kotian, M.S. Graduate Program in Molecular, Cellular and Developmental Biology The Ohio State University 2013 Dissertation Committee: Professor Jeffrey D. Parvin, Advisor Professor Joanna L. Groden Professor Denis C. Guttridge Professor Mark R. Parthun Copyright by Shweta Kotian 2013 Abstract BRCA1 is an important breast- and ovarian-specific tumor suppressor gene. It is important in various cellular functions in the body including transcriptional regulation, cell cycle checkpoint activation, DNA damage response, and maintenance of genomic stability. Approximately 40% of hereditary breast cancers have mutations in BRCA1 or BRCA2, and it is unclear how the remaining cases are caused, presumably by mutations or in the alteration of expression of unknown genes. We hypothesize that these unknown genes or ‘missing BRCAs’ can be identified by bio-informatics methods. Performing gene co-expression analysis, we have compared the expression profiles of hundreds of genes across publicly available breast cancer microarray datasets, with those of BRCA1 and BRCA2. We propose that the genes, whose expression is highly correlated with BRCA1 and BRCA2, might function together in the same pathways. They could also potentially interact with each other. The main aim of this study is to identify these genes, and then biologically validate them in functional BRCA1-regulated pathways including homologous recombination and centrosome duplication. We also hope to find any mechanistic ii associations between these genes and BRCA1. Eventually, we hope to evaluate if these genes contribute to the process of carcinogenesis. As a result of the informatics data mining, we narrowed down the list of genes co- expressed with BRCA1 to a few dozen. Of these genes, we decided to choose the histone deacetylases (HDACs) and NUSAP1 for biological validation. We had several criteria for choosing these, such as 1) the presence of enzymatic domains in the case of the HDAC proteins, 2) an unknown association with BRCA1, 3) a paucity of the literature about this gene/protein; and 4) cancer- associated changes in gene expression. Biological validation was performed using two main assays: homology-directed repair and centrosome duplication. BRCA1 is important for proper DNA repair, as well as regulation of centrosome numbers. When BRCA1 is depleted from cells in these assays using RNA interference, homologous recombination is impaired, and there is aberrant centrosome duplication causing formation of supernumerary centrosomes. Likewise, we were interested in determining if the HDACs or NUSAP1 would be important for these processes as well. The HDAC family catalyzes the deacetylation of histones on chromatin causing chromatin compaction. This is turn, suppresses transcription. In this way, HDACs help regulate the expression of key developmental, cell cycle, and transcription factors. We observed that multiple members of the HDAC family of genes were co-expressed with BRCA1 and BRCA2 in multiple gene expression datasets. Dysregulation of HDACs has been observed in several cancers, and there is iii much interest in the development of HDAC inhibitor drugs. These drugs inhibit the growth of cancer cells, but it is not known how. Thus, we decided to test HDAC function in homologous recombination using these inhibitors. A tissue- culture based homology directed repair (HDR) assay was used in which repair of a double-stranded break, by homologous recombination, results in gene conversion of an inactive GFP allele to an active GFP gene. Homologous recombination events were then readily scored by measuring GFP positive cells by FACS. We found that treatment with HDAC inhibitors significantly reduced homologous recombination levels. Upon carrying out an individual depletion of HDACs 1 to 11 by RNA interference, we found that depletions of HDAC9 and HDAC10 specifically reduce homologous recombination levels. This was corroborated as HDAC9 and HDAC10 depletion sensitized cells to the interstrand crosslinker mitomycin C, indicating faulty homologous recombination. We think that HDAC9 and HDAC10 catalyze a crucial deacetylation step at the site of the double strand break that is important for homologous recombination. NUSAP1 is important for proper mitotic spindle assembly and cytokinesis. It is also aberrantly expressed in several cancers. We performed NUSAP1 depletion in the HDR assay, and saw that homologous recombination was impaired. In addition, depletion of NUSAP1 in the centrosome assay, caused centrosome amplification. Over-expression of BRCA1 reversed the defective phenotypes seen in both these assays upon NUSAP1 depletion. While NUSAP1 depletion reduced BRCA1 protein levels in the soluble extracts, and specifically in the iv chromatin fractions, there was no change in total BRCA1 protein levels. This indicated that NUSAP1 controls BRCA1 localization. We further confirmed this by determining that NUSAP1 depletion inhibited recruitment of BRCA1 to DNA damage-induced foci upon ionizing irradiation. In addition to BRCA1, RAD51 recruitment to the DNA damage foci was affected. Thus, NUSAP1 impacts the DNA damage response through control of BRCA1 localization. Thus, we have successfully identified and validated three novel genes- HDAC9, HDAC10, NUSAP1 as important in the BRCA1-regulated pathway of homologous recombination. NUSAP1 is also important for centrosome amplification. Since, these genes are implicated in cancer; we hope that our study will help shed light on exactly how they are important for tumorigenesis. Further work will determine whether these proteins may be useful as biomarkers for breast cancer. v Dedication I dedicate this document to my grandparents, the late Mr. Rajeeva Suvarna and the late Mrs. Vimala Suvarna. Words cannot describe how truly grateful I am for their loving care and support in my crucial, formative years. vi Acknowledgments I would like to offer my humble gratitude to my advisor Dr. Jeffrey D. Parvin. Not only is he an immense source of knowledge, but he also has immense patience. I am thankful to him for his invaluable guidance, but most of all for not giving up on me during a very difficult phase in my life. Thanks are due to the members of my lab who have always offered help willingly when needed. I would also like to make a special mention of my colleague, Ms. Mansi Arora, for offering both professional and personal support. I would like to sincerely thank my committee members, Dr. Joanna L. Groden, Dr. Mark R. Parthun, and Dr. Denis C. Guttridge, for offering their valuable time and expertise to help guide me with my dissertation. I would also like to thank my program chair, Dr. David Bisaro, for always being willing to lend a listening ear to even a graduate student’s petty problems, and offer practical advice. This dissertation would not be complete without acknowledging some key people in my life. I will always be grateful to my best friend and partner, Mr. Daniel Stanojevic, who has provided tremendous emotional support to me during this journey. Last, but not least, I am eternally indebted to my mother, Ms. Shashikala vii Kotian, whose fortitude and personal sacrifices, have made it possible for me to reach this personal milestone in my life. viii Vita 2004 ....................................................... B.S. Biochemistry, University of Mumbai 2006 ....................................................... M.S. Biochemistry, University of Mumbai 2006 to present ...................................... Graduate Research Associate, Department of Biomedical Informatics, The Ohio State University Publications Kotian S, Parvin JD. NUSAP1 Influences the DNA damage response by controlling BRCA1 localization. Manuscript in preparation Zhang J, Lu K, Xiang Y, Islam M, Kotian S, Kais Z, Lee C, Arora A, Liu H, Parvin JD, Huang K (2012). Weighted frequent gene co-expression network mining to identify genes involved in genome stability. PLoS Comput Biol. 8(8):e1002656. Epub 2012 Aug 30. Kotian S, Liyanarachchi S, Zelent A, Parvin JD (2011). Histone deacetylases 9 and 10 are required for homologous recombination. J Biol Chem, 286(10):7722-6 ix Fields of Study Major Field: Molecular, Cellular and Developmental Biology x Table of Contents Abstract .................................................................................................................ii Dedication ............................................................................................................vi Acknowledgments ............................................................................................... vii Vita .......................................................................................................................ix Publications ..........................................................................................................ix Fields of Study ...................................................................................................... x Table of Contents .................................................................................................xi List of Tables ...................................................................................................... xvi List of Figures .................................................................................................... xvii Chapter 1:
Recommended publications
  • Transcription-Induced DNA Double Strand Breaks: Both Oncogenic Force and Potential Therapeutic Target?
    Published OnlineFirst March 8, 2011; DOI: 10.1158/1078-0432.CCR-10-2044 Clinical Cancer Molecular Pathways Research Transcription-Induced DNA Double Strand Breaks: Both Oncogenic Force and Potential Therapeutic Target? Michael C. Haffner, Angelo M. De Marzo, Alan K. Meeker, William G. Nelson, and Srinivasan Yegnasubramanian Abstract An emerging model of transcriptional activation suggests that induction of transcriptional programs, for instance by stimulating prostate or breast cells with androgens or estrogens, respectively, involves the formation of DNA damage, including DNA double strand breaks (DSB), recruitment of DSB repair proteins, and movement of newly activated genes to transcription hubs. The DSB can be mediated by the class II topoisomerase TOP2B, which is recruited with the androgen receptor and estrogen receptor to regulatory sites on target genes and is apparently required for efficient transcriptional activation of these genes. These DSBs are recognized by the DNA repair machinery triggering the recruitment of repair proteins such as poly(ADP-ribose) polymerase 1 (PARP1), ATM, and DNA-dependent protein kinase (DNA-PK). If illegitimately repaired, such DSBs can seed the formation of genomic rearrangements like the TMPRSS2- ERG fusion oncogene in prostate cancer. Here, we hypothesize that these transcription-induced, TOP2B- mediated DSBs can also be exploited therapeutically and propose that, in hormone-dependent tumors like breast and prostate cancers, a hormone-cycling therapy, in combination with topoisomerase II poisons or inhibitors of the DNA repair components PARP1 and DNA-PK, could overwhelm cancer cells with transcription-associated DSBs. Such strategies may find particular utility in cancers, like prostate cancer, which show low proliferation rates, in which other chemotherapeutic strategies that target rapidly proliferating cells have had limited success.
    [Show full text]
  • Targeting Topoisomerase I in the Era of Precision Medicine Anish Thomas and Yves Pommier
    Published OnlineFirst June 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1089 Review Clinical Cancer Research Targeting Topoisomerase I in the Era of Precision Medicine Anish Thomas and Yves Pommier Abstract Irinotecan and topotecan have been widely used as including the indenoisoquinolines LMP400 (indotecan), anticancer drugs for the past 20 years. Because of their LMP776 (indimitecan), and LMP744, and on tumor- selectivity as topoisomerase I (TOP1) inhibitors that trap targeted delivery TOP1 inhibitors using liposome, PEGyla- TOP1 cleavage complexes, camptothecins are also widely tion, and antibody–drug conjugates. We also address how used to elucidate the DNA repair pathways associated with tumor-specific determinants such as homologous recombi- DNA–protein cross-links and replication stress. This review nation defects (HRD and BRCAness) and Schlafen 11 summarizes the basic molecular mechanisms of action (SLFN11) expression can be used to guide clinical appli- of TOP1 inhibitors, their current use, and limitations cation of TOP1 inhibitors in combination with DNA dam- as anticancer agents. We introduce new therapeutic strate- age response inhibitors including PARP, ATR, CHEK1, and gies based on novel TOP1 inhibitor chemical scaffolds ATM inhibitors. Introduction DNA structures such as plectonemes, guanosine quartets, R-loops, and DNA breaks (reviewed in ref. 1). Humans encodes six topoisomerases, TOP1, TOP1MT, TOP2a, TOP2b, TOP3a, and TOP3b (1) to pack and unpack the approx- imately 2 meters of DNA that needs to be contained in the nucleus Anticancer TOP1 Inhibitors Trap TOP1CCs whose diameter (6 mm) is approximately 3 million times smaller. as Interfacial Inhibitors Moreover, the genome is organized in chromosome loops and the separation of the two strands of DNA during transcription and The plant alkaloid camptothecin and its clinical derivatives, replication generate torsional stress and supercoils that are topotecan and irinotecan (Fig.
    [Show full text]
  • Single-Nuclei RNA-Seq on Human Retinal Tissue Provides Improved Transcriptome Profiling
    ARTICLE https://doi.org/10.1038/s41467-019-12917-9 OPEN Single-nuclei RNA-seq on human retinal tissue provides improved transcriptome profiling Qingnan Liang 1,2,3,9, Rachayata Dharmat1,2,8,9, Leah Owen4, Akbar Shakoor4, Yumei Li1, Sangbae Kim1, Albert Vitale4, Ivana Kim4, Denise Morgan4,5, Shaoheng Liang 6, Nathaniel Wu1, Ken Chen 6, Margaret M. DeAngelis4,5,7* & Rui Chen1,2,3* Single-cell RNA-seq is a powerful tool in decoding the heterogeneity in complex tissues by 1234567890():,; generating transcriptomic profiles of the individual cell. Here, we report a single-nuclei RNA- seq (snRNA-seq) transcriptomic study on human retinal tissue, which is composed of mul- tiple cell types with distinct functions. Six samples from three healthy donors are profiled and high-quality RNA-seq data is obtained for 5873 single nuclei. All major retinal cell types are observed and marker genes for each cell type are identified. The gene expression of the macular and peripheral retina is compared to each other at cell-type level. Furthermore, our dataset shows an improved power for prioritizing genes associated with human retinal dis- eases compared to both mouse single-cell RNA-seq and human bulk RNA-seq results. In conclusion, we demonstrate that obtaining single cell transcriptomes from human frozen tissues can provide insight missed by either human bulk RNA-seq or animal models. 1 HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA. 2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston 77030 TX, USA. 3 Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Download Download
    Supplementary Figure S1. Results of flow cytometry analysis, performed to estimate CD34 positivity, after immunomagnetic separation in two different experiments. As monoclonal antibody for labeling the sample, the fluorescein isothiocyanate (FITC)- conjugated mouse anti-human CD34 MoAb (Mylteni) was used. Briefly, cell samples were incubated in the presence of the indicated MoAbs, at the proper dilution, in PBS containing 5% FCS and 1% Fc receptor (FcR) blocking reagent (Miltenyi) for 30 min at 4 C. Cells were then washed twice, resuspended with PBS and analyzed by a Coulter Epics XL (Coulter Electronics Inc., Hialeah, FL, USA) flow cytometer. only use Non-commercial 1 Supplementary Table S1. Complete list of the datasets used in this study and their sources. GEO Total samples Geo selected GEO accession of used Platform Reference series in series samples samples GSM142565 GSM142566 GSM142567 GSM142568 GSE6146 HG-U133A 14 8 - GSM142569 GSM142571 GSM142572 GSM142574 GSM51391 GSM51392 GSE2666 HG-U133A 36 4 1 GSM51393 GSM51394 only GSM321583 GSE12803 HG-U133A 20 3 GSM321584 2 GSM321585 use Promyelocytes_1 Promyelocytes_2 Promyelocytes_3 Promyelocytes_4 HG-U133A 8 8 3 GSE64282 Promyelocytes_5 Promyelocytes_6 Promyelocytes_7 Promyelocytes_8 Non-commercial 2 Supplementary Table S2. Chromosomal regions up-regulated in CD34+ samples as identified by the LAP procedure with the two-class statistics coded in the PREDA R package and an FDR threshold of 0.5. Functional enrichment analysis has been performed using DAVID (http://david.abcc.ncifcrf.gov/)
    [Show full text]
  • The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression
    International Journal of Molecular Sciences Review The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression Tingting Zou and Zhenghong Lin * School of Life Sciences, Chongqing University, Chongqing 401331, China; [email protected] * Correspondence: [email protected] Abstract: The cell cycle is a collection of events by which cellular components such as genetic materials and cytoplasmic components are accurately divided into two daughter cells. The cell cycle transition is primarily driven by the activation of cyclin-dependent kinases (CDKs), which activities are regulated by the ubiquitin-mediated proteolysis of key regulators such as cyclins, CDK inhibitors (CKIs), other kinases and phosphatases. Thus, the ubiquitin-proteasome system (UPS) plays a pivotal role in the regulation of the cell cycle progression via recognition, interaction, and ubiquitination or deubiquitination of key proteins. The illegitimate degradation of tumor suppressor or abnormally high accumulation of oncoproteins often results in deregulation of cell proliferation, genomic instability, and cancer occurrence. In this review, we demonstrate the diversity and complexity of the regulation of UPS machinery of the cell cycle. A profound understanding of the ubiquitination machinery will provide new insights into the regulation of the cell cycle transition, cancer treatment, and the development of anti-cancer drugs. Keywords: cell cycle regulation; CDKs; cyclins; CKIs; UPS; E3 ubiquitin ligases; Deubiquitinases (DUBs) Citation: Zou, T.; Lin, Z. The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression. 1. Introduction Int. J. Mol. Sci. 2021, 22, 5754. https://doi.org/10.3390/ijms22115754 The cell cycle is a ubiquitous, complex, and highly regulated process that is involved in the sequential events during which a cell duplicates its genetic materials, grows, and di- Academic Editors: Kwang-Hyun Bae vides into two daughter cells.
    [Show full text]
  • The APC/C in Female Mammalian Meiosis I
    REPRODUCTIONREVIEW The APC/C in female mammalian meiosis I Hayden Homer1,2 1Mammalian Oocyte and Embryo Research Laboratory, Cell and Developmental Biology, UCL, London WC1E 6BT, UK and 2Reproductive Medicine Unit, Institute for Women’s Health, UCLH Elizabeth Garrett Anderson Wing, London NW1 2BU, UK Correspondence should be addressed to H Homer; Email: [email protected] Abstract The anaphase-promoting complex or cyclosome (APC/C) orchestrates a meticulously controlled sequence of proteolytic events critical for proper cell cycle progression, the details of which have been most extensively elucidated during mitosis. It has become apparent, however, that the APC/C, particularly when acting in concert with its Cdh1 co-activator (APC/CCdh1), executes a staggeringly diverse repertoire of functions that extend its remit well outside the bounds of mitosis. Findings over the past decade have not only earmarked mammalian oocyte maturation as one such case in point but have also begun to reveal a complex pattern of APC/C regulation that underpins many of the oocyte’s unique developmental attributes. This review will encompass the latest findings pertinent to the APC/C, especially APC/CCdh1, in mammalian oocytes and how its activity and substrates shape the stop–start tempo of female mammalian first meiotic division and the challenging requirement for assembling spindles in the absence of centrosomes. Reproduction (2013) 146 R61–R71 Introduction associated somatic follicular compartment at the time of ovulation. Significantly, although primordial germ cells Meiosis is the unique cell division that halves the in the ovary commit to meiosis during fetal life, it is chromosome compliment by coupling two successive not until postnatal adulthood that mature oocytes (or nuclear divisions with a single round of DNA replication.
    [Show full text]
  • 1 Spindle Assembly Checkpoint Is Sufficient for Complete Cdc20
    Spindle assembly checkpoint is sufficient for complete Cdc20 sequestering in mitotic control Bashar Ibrahim Bio System Analysis Group, Friedrich-Schiller-University Jena, and Jena Centre for Bioinformatics (JCB), 07743 Jena, Germany Email: [email protected] Abstract The spindle checkpoint assembly (SAC) ensures genome fidelity by temporarily delaying anaphase onset, until all chromosomes are properly attached to the mitotic spindle. The SAC delays mitotic progression by preventing activation of the ubiquitin ligase anaphase-promoting complex (APC/C) or cyclosome; whose activation by Cdc20 is required for sister-chromatid separation marking the transition into anaphase. The mitotic checkpoint complex (MCC), which contains Cdc20 as a subunit, binds stably to the APC/C. Compelling evidence by Izawa and Pines (Nature 2014; 10.1038/nature13911) indicates that the MCC can inhibit a second Cdc20 that has already bound and activated the APC/C. Whether or not MCC per se is sufficient to fully sequester Cdc20 and inhibit APC/C remains unclear. Here, a dynamic model for SAC regulation in which the MCC binds a second Cdc20 was constructed. This model is compared to the MCC, and the MCC-and-BubR1 (dual inhibition of APC) core model variants and subsequently validated with experimental data from the literature. By using ordinary nonlinear differential equations and spatial simulations, it is shown that the SAC works sufficiently to fully sequester Cdc20 and completely inhibit APC/C activity. This study highlights the principle that a systems biology approach is vital for molecular biology and could also be used for creating hypotheses to design future experiments. Keywords: Mathematical biology, Spindle assembly checkpoint; anaphase promoting complex, MCC, Cdc20, systems biology 1 Introduction Faithful DNA segregation, prior to cell division at mitosis, is vital for maintaining genomic integrity.
    [Show full text]
  • Bub1 Positions Mad1 Close to KNL1 MELT Repeats to Promote Checkpoint Signalling
    ARTICLE Received 14 Dec 2016 | Accepted 3 May 2017 | Published 12 June 2017 DOI: 10.1038/ncomms15822 OPEN Bub1 positions Mad1 close to KNL1 MELT repeats to promote checkpoint signalling Gang Zhang1, Thomas Kruse1, Blanca Lo´pez-Me´ndez1, Kathrine Beck Sylvestersen1, Dimitriya H. Garvanska1, Simone Schopper1, Michael Lund Nielsen1 & Jakob Nilsson1 Proper segregation of chromosomes depends on a functional spindle assembly checkpoint (SAC) and requires kinetochore localization of the Bub1 and Mad1/Mad2 checkpoint proteins. Several aspects of Mad1/Mad2 kinetochore recruitment in human cells are unclear and in particular the underlying direct interactions. Here we show that conserved domain 1 (CD1) in human Bub1 binds directly to Mad1 and a phosphorylation site exists in CD1 that stimulates Mad1 binding and SAC signalling. Importantly, fusion of minimal kinetochore-targeting Bub1 fragments to Mad1 bypasses the need for CD1, revealing that the main function of Bub1 is to position Mad1 close to KNL1 MELTrepeats. Furthermore, we identify residues in Mad1 that are critical for Mad1 functionality, but not Bub1 binding, arguing for a direct role of Mad1 in the checkpoint. This work dissects functionally relevant molecular interactions required for spindle assembly checkpoint signalling at kinetochores in human cells. 1 The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark. Correspondence and requests for materials should be addressed to G.Z.
    [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]
  • The Role of Sumoylation of DNA Topoisomerase Iiα C-Terminal Domain in the Regulation of Mitotic Kinases In
    SUMOylation at the centromere: The role of SUMOylation of DNA topoisomerase IIα C-terminal domain in the regulation of mitotic kinases in cell cycle progression. By Makoto Michael Yoshida Submitted to the graduate degree program in the Department of Molecular Biosciences and the Graduate Faculty of the University of Kansas in partial fulfillment of the requirements for the degree of Doctor of Philosophy. ________________________________________ Chairperson: Yoshiaki Azuma, Ph.D. ________________________________________ Roberto De Guzman, Ph.D. ________________________________________ Kristi Neufeld, Ph.D. _________________________________________ Berl Oakley, Ph.D. _________________________________________ Blake Peterson, Ph.D. Date Defended: July 12, 2016 The Dissertation Committee for Makoto Michael Yoshida certifies that this is the approved version of the following dissertation: SUMOylation at the centromere: The role of SUMOylation of DNA topoisomerase IIα C-terminal domain in the regulation of mitotic kinases in cell cycle progression. ________________________________________ Chairperson: Yoshiaki Azuma, Ph.D. Date approved: July 12, 2016 ii ABSTRACT In many model systems, SUMOylation is required for proper mitosis; in particular, chromosome segregation during anaphase. It was previously shown that interruption of SUMOylation through the addition of the dominant negative E2 SUMO conjugating enzyme Ubc9 in mitosis causes abnormal chromosome segregation in Xenopus laevis egg extract (XEE) cell-free assays, and DNA topoisomerase IIα (TOP2A) was identified as a substrate for SUMOylation at the mitotic centromeres. TOP2A is SUMOylated at K660 and multiple sites in the C-terminal domain (CTD). We sought to understand the role of TOP2A SUMOylation at the mitotic centromeres by identifying specific binding proteins for SUMOylated TOP2A CTD. Through affinity isolation, we have identified Haspin, a histone H3 threonine 3 (H3T3) kinase, as a SUMOylated TOP2A CTD binding protein.
    [Show full text]
  • Differential Expression of Multiple Disease-Related Protein Groups
    brain sciences Article Differential Expression of Multiple Disease-Related Protein Groups Induced by Valproic Acid in Human SH-SY5Y Neuroblastoma Cells 1,2, 1, 1 1 Tsung-Ming Hu y, Hsiang-Sheng Chung y, Lieh-Yung Ping , Shih-Hsin Hsu , Hsin-Yao Tsai 1, Shaw-Ji Chen 3,4 and Min-Chih Cheng 1,* 1 Department of Psychiatry, Yuli Branch, Taipei Veterans General Hospital, Hualien 98142, Taiwan; [email protected] (T.-M.H.); [email protected] (H.-S.C.); [email protected] (L.-Y.P.); fi[email protected] (S.-H.H.); [email protected] (H.-Y.T.) 2 Department of Future Studies and LOHAS Industry, Fo Guang University, Jiaosi, Yilan County 26247, Taiwan 3 Department of Psychiatry, Mackay Medical College, New Taipei City 25245, Taiwan; [email protected] 4 Department of Psychiatry, Taitung Mackay Memorial Hospital, Taitung County 95064, Taiwan * Correspondence: [email protected]; Tel.: +886-3888-3141 (ext. 475) These authors contributed equally to this work. y Received: 10 July 2020; Accepted: 8 August 2020; Published: 12 August 2020 Abstract: Valproic acid (VPA) is a multifunctional medication used for the treatment of epilepsy, mania associated with bipolar disorder, and migraine. The pharmacological effects of VPA involve a variety of neurotransmitter and cell signaling systems, but the molecular mechanisms underlying its clinical efficacy is to date largely unknown. In this study, we used the isobaric tags for relative and absolute quantitation shotgun proteomic analysis to screen differentially expressed proteins in VPA-treated SH-SY5Y cells. We identified changes in the expression levels of multiple proteins involved in Alzheimer’s disease, Parkinson’s disease, chromatin remodeling, controlling gene expression via the vitamin D receptor, ribosome biogenesis, ubiquitin-mediated proteolysis, and the mitochondrial oxidative phosphorylation and electron transport chain.
    [Show full text]