23 results on '"Jacqueline J.L. Jacobs"'
Search Results
2. REV7: Jack of many trades
- Author
-
Jacqueline J.L. Jacobs, Vera Boersma, and Inge de Krijger
- Subjects
0303 health sciences ,DNA Repair ,Genome integrity ,Cell Cycle Proteins ,DNA ,Cell Biology ,HORMA domain ,Computational biology ,Mitotic progression ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Genome maintenance ,Mad2 Proteins ,Humans ,DNA Breaks, Double-Stranded ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
The HORMA domain protein REV7, also known as MAD2L2, interacts with a variety of proteins and thereby contributes to the establishment of different complexes. With doing so, REV7 impacts a diverse range of cellular processes and gained increasing interest as more of its activities became uncovered. REV7 has important roles in translesion synthesis and mitotic progression, and acts as a central component in the recently discovered shieldin complex that operates in DNA double-strand break repair. Here we discuss the roles of REV7 in its various complexes, focusing on its activity in genome integrity maintenance. Moreover, we will describe current insights on REV7 structural features that allow it to be such a versatile protein.
- Published
- 2021
3. Shieldin complex promotes DNA end-joining and counters homologous recombination in BRCA1-null cells
- Author
-
Stephen P. Jackson, Mareike Herzog, Alejandra Bruna, Luca Pellegrini, Violeta Serra, Mark J. O'Connor, Zhongwu Lai, Chloé Lescale, Jacqueline J.L. Jacobs, Fengtang Yang, Jonathan Lam, Matylda Sczaniecka-Clift, Abigail Shea, Carlos Caldas, Matthias Ostermaier, Gabriel Balmus, Julia Coates, Wenming Wei, Inge de Krijger, Yaron Galanty, Mukerrem Demir, Ludovic Deriano, Petra Beli, Domenic Pilger, Harveer Dev, Rimma Belotserkovskaya, Alistair Martin, Beiyuan Fu, Ting-Wei Will Chiang, Qian Wu, Dev, Harveer [0000-0003-2874-6894], Yang, Fengtang [0000-0002-3573-2354], Balmus, Gabriel [0000-0003-2872-4468], Serra, Violeta [0000-0001-6620-1065], Beli, Petra [0000-0001-9507-9820], Pellegrini, Luca [0000-0002-9300-497X], Deriano, Ludovic [0000-0002-9673-9525], Jacobs, Jacqueline JL [0000-0002-7704-4795], Jackson, Stephen P [0000-0001-9317-7937], Apollo - University of Cambridge Repository, Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge [UK] (CAM), Department of Biochemistry [Cambridge], Cambridge University Hospitals - NHS (CUH), Intégrité du génome, immunité et cancer - Genome integrity, Immunity and Cancer, Institut Pasteur [Paris] (IP), Netherlands Cancer Institute (NKI), Antoni van Leeuwenhoek Hospital, Cancer Research UK Cambridge Institute [Cambridge, Royaume-Uni] (CRUK), Institute of Molecular Biology (IMB), Johannes Gutenberg - Universität Mainz = Johannes Gutenberg University (JGU), Wellcome Trust Sanger Institute [Hinxton, UK], AstraZeneca US [Waltham, USA], AstraZeneca [Cambridge, UK], The Wellcome Trust Sanger Institute [Cambridge], Vall d'Hebron Institute of Oncology [Barcelone] (VHIO), Vall d'Hebron University Hospital [Barcelona], The SPJ lab is largely funded by a Cancer Research UK (CRUK) Program Grant, C6/A18796, and a Wellcome Trust (WT) Investigator Award, 206388/Z/17/Z. Core infrastructure funding was provided by CRUK grant C6946/A24843 and WT grant WT203144. S.P.J. receives a salary from the University of Cambridge. H.D. is funded by WT Clinical Fellowship 206721/Z/17/Z. TWC was supported by a Cambridge International Scholarship. D.P. is funded by Cancer Research UK studentship C6/A21454. The P.B. lab is supported by the Emmy Noether Program (BE 5342/1-1) from the German Research Foundation and a Marie Curie Career Integration Grant from the European Commission (630763). The L.P. lab is funded by the WT (investigator award 104641/Z/14/Z) and the Medical Research Council (project grant MR/N000161/1). The C.C. lab was supported with funding from CRUK. The J.J. lab was supported by the European Research Council grant ERC-StG 311565, The Dutch Cancer Society (KWF) grant KWF 10999, and the Netherlands Organization for Scientific Research (NWO) as part of the National Roadmap Large-scale Research Facilities of the Netherlands, Proteins@Work (project no. 184.032.201 to the Proteomics Facility of the Netherlands Cancer Institute). The L.D. lab is funded by the Institut Pasteur, the Institut National du Cancer (no. PLBIO16-181) and the European Research Council (starting grant agreement no. 310917). W.W. is part of the Pasteur–Paris University (PPU) International PhD program and this project received funding from the CNBG company, China. Q.W. is funded by the Wellcome Trust (200814/Z/16/Z ). The V.S. lab work was funded by the Instituto de Salud Carlos III (ISCIII), an initiative of the Spanish Ministry of Economy and Innovation partially supported by European Regional Development FEDER Funds (PI17-01080 to VS), the European Research Area-NET, Transcan-2 (AC15/00063), a non-commercial research agreement with AstraZeneca UK, and structural funds from the Agència de Gestió d’Ajuts Universitaris i de Recerca (AGAUR, 2017 SGR 540) and the Orozco Family. V.S. received a salary and travel support to C.C.’s lab from ISCIII (CP14/00228, MV15/00041) and the FERO Foundation., The authors thank all S.P.J. laboratory members for support and advice, and Cambridge colleagues N. Lawrence for OMX super-resolution microscopy support and R. Butler for help with computational image analyses and programming. The authors also thank S. Selivanova and S. Hough for help with plasmid amplification, sample preparation and tissue culture maintenance, K. Dry for extensive editorial assistance, F. Muñoz-Martinez for assistance with CRISPR–Cas9 knockout generation, L. Radu for assistance with protein purification, C. Lord (Institute of Cancer Research, London) for SUM149PT cells, D. Durocher (University of Toronto, Canada) for U2OS LacSceIII cells, F. Alt (Harvard University, USA) for CH12F3 cells and 53bp1 knockout CH12F3 cell clones, T. Honjo (Kyoto University, Japan) for permission to use the CH12F3 cell line, and J. Serrat in the Jacobs lab for technical assistance, Institut Pasteur [Paris], and Johannes Gutenberg - Universität Mainz (JGU)
- Subjects
MESH: DNA Breaks, Double-Stranded ,RAD51 ,Cell Cycle Proteins ,Poly (ADP-Ribose) Polymerase Inhibitor ,MESH: Recombinational DNA Repair ,Mice ,MESH: Animals ,DNA Breaks, Double-Stranded ,skin and connective tissue diseases ,Cancer ,Telomere-binding protein ,Ovarian Neoplasms ,MESH: Breast Neoplasms / metabolism ,MESH: Telomere-Binding Proteins / metabolism ,3. Good health ,Cell biology ,MESH: HEK293 Cells ,MESH: Proteins / genetics ,MESH: Telomere-Binding Proteins / genetics ,MESH: Tumor Suppressor p53-Binding Protein 1 / metabolism ,MESH: Xenograft Model Antitumor Assays ,Telomere-Binding Proteins ,MESH: Ovarian Neoplasms / drug therapy ,Bone Neoplasms ,MESH: Ovarian Neoplasms / metabolism ,Article ,03 medical and health sciences ,MESH: Cell Cycle Proteins ,MESH: Bone Neoplasms / metabolism ,Humans ,MESH: Osteosarcoma / metabolism ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,MESH: Humans ,MESH: Tumor Suppressor p53-Binding Protein 1 / genetics ,Dose-Response Relationship, Drug ,HEK 293 cells ,Proteins ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,DNA ,MESH: BRCA1 Protein / deficiency ,030104 developmental biology ,Multiprotein Complexes ,MESH: Mad2 Proteins / metabolism ,MESH: Breast Neoplasms / genetics ,MESH: Bone Neoplasms / drug therapy ,Cisplatin ,Homologous recombination ,MESH: Osteosarcoma / genetics ,MESH: Female ,0301 basic medicine ,DNA End-Joining Repair ,MESH: Proteins / metabolism ,MESH: Dose-Response Relationship, Drug ,chemistry.chemical_compound ,MESH: Osteosarcoma / pathology ,MESH: Breast Neoplasms / pathology ,Homologous Recombination ,Polymerase ,MESH: Breast Neoplasms / drug therapy ,Osteosarcoma ,biology ,Chemistry ,BRCA1 Protein ,DNA damage and repair ,MESH: Poly(ADP-ribose) Polymerase Inhibitors / pharmacology ,MESH: Bone Neoplasms / genetics ,DNA-Binding Proteins ,MESH: Bone Neoplasms / pathology ,Mad2 Proteins ,Female ,MESH: Ovarian Neoplasms / genetics ,Tumor Suppressor p53-Binding Protein 1 ,MESH: Cisplatin / pharmacology ,MESH: Cell Line, Tumor ,Lymphocytes, Null ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Breast Neoplasms ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,MESH: BRCA1 Protein / genetics ,Poly(ADP-ribose) Polymerase Inhibitors ,Cell Line, Tumor ,MESH: Drug Resistance, Neoplasm* / genetics ,MESH: Mad2 Proteins / genetics ,MESH: Ovarian Neoplasms / pathology ,Animals ,MESH: Mice ,MESH: Osteosarcoma / drug therapy ,Oligonucleotide ,Protective Devices ,Recombinational DNA Repair ,Cell Biology ,MESH: Multiprotein Complexes ,Xenograft Model Antitumor Assays ,HEK293 Cells ,Drug Resistance, Neoplasm ,biology.protein ,MESH: DNA End-Joining Repair ,MESH: DNA-Binding Proteins - Abstract
International audience; BRCA1 deficiencies cause breast, ovarian, prostate and other cancers, and render tumours hypersensitive to poly(ADP-ribose) polymerase (PARP) inhibitors. To understand the resistance mechanisms, we conducted whole-genome CRISPR-Cas9 synthetic-viability/resistance screens in BRCA1-deficient breast cancer cells treated with PARP inhibitors. We identified two previously uncharacterized proteins, C20orf196 and FAM35A, whose inactivation confers strong PARP-inhibitor resistance. Mechanistically, we show that C20orf196 and FAM35A form a complex, 'Shieldin' (SHLD1/2), with FAM35A interacting with single-stranded DNA through its C-terminal oligonucleotide/oligosaccharide-binding fold region. We establish that Shieldin acts as the downstream effector of 53BP1/RIF1/MAD2L2 to promote DNA double-strand break (DSB) end-joining by restricting DSB resection and to counteract homologous recombination by antagonizing BRCA2/RAD51 loading in BRCA1-deficient cells. Notably, Shieldin inactivation further sensitizes BRCA1-deficient cells to cisplatin, suggesting how defining the SHLD1/2 status of BRCA1-deficient tumours might aid patient stratification and yield new treatment opportunities. Highlighting this potential, we document reduced SHLD1/2 expression in human breast cancers displaying intrinsic or acquired PARP-inhibitor resistance.
- Published
- 2018
4. The CST complex mediates end protection at double-strand breaks and promotes PARP inhibitor sensitivity in BRCA1-deficient cells
- Author
-
Stefano Annunziato, Jinhyuk Bhin, Marieke van de Ven, Rachel Brough, Bastiaan Evers, Marco Barazas, Catrin Lutz, Stephen J. Pettitt, Dik C. van Gent, Jacqueline J.L. Jacobs, Stefan J. Roobol, Jessica Frankum, Jos Jonkers, Inge de Krijger, Hind Ghezraoui, Fei Fei Song, Christopher J. Lord, J. Ross Chapman, Ewa Gogola, Sven Rottenberg, Molecular Genetics, and Radiology & Nuclear Medicine
- Subjects
0301 basic medicine ,DNA end resection ,Poly ADP ribose polymerase ,610 Medicine & health ,Synthetic lethality ,CST complex ,Poly(ADP-ribose) Polymerase Inhibitors ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,breast cancer ,Cell Line, Tumor ,Animals ,DNA Breaks, Double-Stranded ,Polymerase ,drug resistance ,biology ,630 Agriculture ,Chemistry ,TEN1 ,BRCA1 Protein ,Mouse Embryonic Stem Cells ,Telomere ,BRCA1 ,genetically engineered mouse model ,3. Good health ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,PARP inhibitor ,STN1 ,Drug Resistance, Neoplasm ,Multiprotein Complexes ,biology.protein ,570 Life sciences ,CTC1 ,Female ,CRISPR-Cas Systems ,Homologous recombination ,DNA - Abstract
Summary Selective elimination of BRCA1-deficient cells by inhibitors of poly(ADP-ribose) polymerase (PARP) is a prime example of the concept of synthetic lethality in cancer therapy. This interaction is counteracted by the restoration of BRCA1-independent homologous recombination through loss of factors such as 53BP1, RIF1, and REV7/MAD2L2, which inhibit end resection of DNA double-strand breaks (DSBs). To identify additional factors involved in this process, we performed CRISPR/SpCas9-based loss-of-function screens and selected for factors that confer PARP inhibitor (PARPi) resistance in BRCA1-deficient cells. Loss of members of the CTC1-STN1-TEN1 (CST) complex were found to cause PARPi resistance in BRCA1-deficient cells in vitro and in vivo. We show that CTC1 depletion results in the restoration of end resection and that the CST complex may act downstream of 53BP1/RIF1. These data suggest that, in addition to its role in protecting telomeres, the CST complex also contributes to protecting DSBs from end resection., Graphical Abstract, Highlights • PARP inhibitor resistance screens independently identify loss of CTC1 as major hit • The CST complex promotes PARP inhibitor sensitivity in BRCA1-deficient cells • Depletion of CTC1 restores end resection in BRCA1-deficient cells • CTC1 facilitates double-strand break repair via canonical non-homologous end joining, Using CRISPR/SpCas9-based loss-of-function screens, Barazas et al. show that loss of the CTC1-STN1-TEN1 (CST) complex promotes PARP inhibitor resistance in BRCA1-deficient cells. Mechanistically, the CST complex maintains double-strand break end stability in addition to its role in protecting telomeric ends.
- Published
- 2018
5. MAD2L2 controls DNA repair at telomeres and DNA breaks by inhibiting 5′ end resection
- Author
-
Vera Boersma, Jacqueline J.L. Jacobs, Daniel Durocher, Alexandre Orthwein, Jaco van der Torre, Marieke H. Peuscher, Sandra Segura-Bayona, Nathalie Moatti, and Brigitte A. Wevers
- Subjects
DNA End-Joining Repair ,Telomere-binding protein ,Genome instability ,Multidisciplinary ,DNA repair ,DNA replication ,REV1 ,Biology ,Molecular biology ,Article ,Nucleotide excision repair ,Cell biology ,Telomere - Abstract
Appropriate repair of DNA lesions and the inhibition of DNA repair activities at telomeres are critical to prevent genomic instability. By fuelling the generation of genetic alterations and by compromising cell viability, genomic instability is a driving force in cancer and aging1, 2. Here we identify MAD2L2 (also known as MAD2B or REV7) through functional genetic screening as a novel factor controlling DNA repair activities at mammalian telomeres. We show that MAD2L2 accumulates at uncapped telomeres and promotes non-homologous end-joining (NHEJ)-mediated fusion of deprotected chromosome ends and genomic instability. MAD2L2 depletion causes elongated 3′ telomeric overhangs, implying that MAD2L2 inhibits 5′ end-resection. End-resection blocks NHEJ while committing to homology-directed repair (HDR) and is under control of 53BP1, RIF1 and PTIP3. Consistent with MAD2L2 promoting NHEJ-mediated telomere fusion by inhibiting 5′ end-resection, knockdown of the nucleases CTIP or EXO1 partially restores telomere-driven genomic instability in MAD2L2-depleted cells. Control of DNA repair by MAD2L2 is not limited to telomeres. MAD2L2 also accumulates and inhibits end-resection at irradiation (IR)-induced DNA double-strand breaks (DSBs) and promotes end-joining of DSBs in multiple settings, including during immunoglobulin class switch recombination (CSR). These activities of MAD2L2 depend on ATM kinase activity, RNF8, RNF168, 53BP1 and RIF1, but not on PTIP, REV1 and REV3, the latter two acting with MAD2L2 in translesion synthesis (TLS)4. Together our data establish MAD2L2 as a critical contributor to the control of DNA repair activity by 53BP1 that promotes NHEJ by inhibiting 5′ end-resection downstream of RIF1.
- Published
- 2015
6. Ubiquitination and SUMOylation in Telomere Maintenance and Dysfunction
- Author
-
Jacqueline J.L. Jacobs, Zeliha Yalçin, and Carolin Selenz
- Subjects
0301 basic medicine ,Genome instability ,Telomerase ,Cell division ,lcsh:QH426-470 ,DNA repair ,DNA damage ,Review ,Biology ,telomerase ,03 medical and health sciences ,ubiquitin ,Genetics ,telomere maintenance ,Genetics (clinical) ,Telomere-binding protein ,Shelterin ,Telomere ,Cell biology ,lcsh:Genetics ,030104 developmental biology ,telomere dysfunction ,SUMO ,Molecular Medicine ,shelterin - Abstract
Telomeres are essential nucleoprotein structures at linear chromosomes that maintain genome integrity by protecting chromosome ends from being recognized and processed as damaged DNA. In addition, they limit the cell’s proliferative capacity, as progressive loss of telomeric DNA during successive rounds of cell division eventually causes a state of telomere dysfunction that prevents further cell division. When telomeres become critically short, the cell elicits a DNA damage response resulting in senescence, apoptosis or genomic instability, thereby impacting on aging and tumorigenesis. Over the past years substantial progress has been made in understanding the role of post-translational modifications in telomere-related processes, including telomere maintenance, replication and dysfunction. This review will focus on recent findings that establish an essential role for ubiquitination and SUMOylation at telomeres.
- Published
- 2017
7. PARP1 Links CHD2-Mediated Chromatin Expansion and H3.3 Deposition to DNA Repair by Non-homologous End-Joining
- Author
-
Martijn S. Luijsterburg, Wouter W. Wiegant, Alex Pines, Rashmi G. Shah, Anton J.L. de Groot, Inge de Krijger, Alfred C.O. Vertegaal, Girish M. Shah, Haico van Attikum, Jacqueline J.L. Jacobs, and Godelieve Smeenk
- Subjects
0301 basic medicine ,DNA End-Joining Repair ,DNA repair ,Poly (ADP-Ribose) Polymerase-1 ,Solenoid (DNA) ,Biology ,Chromatin remodeling ,Article ,Genomic Instability ,Histones ,03 medical and health sciences ,Cell Line, Tumor ,Histone H2A ,Histone code ,Humans ,DNA Breaks, Double-Stranded ,Molecular Biology ,fungi ,Cell Biology ,Chromatin Assembly and Disassembly ,Molecular biology ,Chromatin ,3. Good health ,Cell biology ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Histone ,HEK293 Cells ,biology.protein ,Poly(ADP-ribose) Polymerases - Abstract
Summary The response to DNA double-strand breaks (DSBs) requires alterations in chromatin structure to promote the assembly of repair complexes on broken chromosomes. Non-homologous end-joining (NHEJ) is the dominant DSB repair pathway in human cells, but our understanding of how it operates in chromatin is limited. Here, we define a mechanism that plays a crucial role in regulating NHEJ in chromatin. This mechanism is initiated by DNA damage-associated poly(ADP-ribose) polymerase 1 (PARP1), which recruits the chromatin remodeler CHD2 through a poly(ADP-ribose)-binding domain. CHD2 in turn triggers rapid chromatin expansion and the deposition of histone variant H3.3 at sites of DNA damage. Importantly, we find that PARP1, CHD2, and H3.3 regulate the assembly of NHEJ complexes at broken chromosomes to promote efficient DNA repair. Together, these findings reveal a PARP1-dependent process that couples ATP-dependent chromatin remodeling with histone variant deposition at DSBs to facilitate NHEJ and safeguard genomic stability., Graphical Abstract, Highlights • PARP1 recruits the chromatin remodeler CHD2 to DNA damage • CHD2 promotes chromatin expansion and H3.3 deposition at DNA breaks • CHD2 promotes the assembly of NHEJ repair complexes at DNA breaks • PARP1 drives CHD2- and H3.3-dependent DNA repair by NHEJ, Luijsterburg et al. define a PARP1-dependent mechanism that regulates NHEJ through localized chromatin expansion and deposition of the histone variant H3.3 by the nucleosome remodeler CHD2 at DNA breaks. Their data also show that these CHD2-mediated events promote DNA repair by facilitating the assembly of NHEJ complexes in chromatin.
- Published
- 2016
8. Posttranslational control of telomere maintenance and the telomere damage response
- Author
-
Jacqueline J.L. Jacobs and Marieke H. Peuscher
- Subjects
Senescence ,Genome instability ,Genetics ,Telomere-binding protein ,DNA Repair ,DNA repair ,Ubiquitination ,Sumoylation ,Cell Biology ,Telomere ,Biology ,Chromatin ,Genomic Instability ,chemistry.chemical_compound ,chemistry ,Humans ,Protein Processing, Post-Translational ,Molecular Biology ,DNA ,Uncapping ,DNA Damage ,Developmental Biology - Abstract
Telomeres help maintain genome integrity by protecting natural chromosome ends from being recognized as damaged DNA. When telomeres become dysfunctional, they limit replicative lifespan and prevent outgrowth of potentially cancerous cells by activating a DNA damage response that forces cells into senescence or apoptosis. On the other hand, chromosome ends devoid of proper telomere protection are subject to DNA repair activities that cause end-to-end fusions and, when cells divide, extensive genomic instability that can promote cancer. While telomeres represent unique chromatin structures with important roles in cancer and aging, we have limited understanding of the way telomeres and the response to their malfunction are controlled at the level of chromatin. Accumulating evidence indicates that different types of posttranslational modifications act in both telomere maintenance and the response to telomere uncapping. Here, we discuss the latest insights on posttranslational control of telomeric chromatin, with emphasis on ubiquitylation and SUMOylation events.
- Published
- 2012
9. Fusing telomeres with RNF8
- Author
-
Jacqueline J.L. Jacobs
- Subjects
Genetics ,Genome instability ,Mutation ,Genome ,DNA repair ,DNA damage ,Ubiquitin-Protein Ligases ,Extra View ,Chromosome ,Cell Biology ,Telomere ,Biology ,medicine.disease_cause ,chemistry.chemical_compound ,chemistry ,Chromosomal Instability ,Chromosome instability ,medicine ,Animals ,Humans ,DNA ,DNA Damage - Abstract
DNA repair activities at DNA double-strand breaks (DSBs) are under control of regulatory ubiquitylation events governed by the RNF8 and RNF168 ubiquitin-ligases. Defects in this regulatory mechanism, as with mutation of other key DNA damage-response factors, lead to genomic instability and cancer, presumably due to impaired repair of DNA lesions. Recent work revealed that RNF8 and RNF168 also play critical roles at natural chromosome ends, when no longer adequately shielded by telomeres. In contrast to repair of DSBs being needed to maintain genome integrity, repair activities at telomeres create chromosome end-to-end fusions that threaten genome integrity. Upon cell division these telomere fusions give rise to genomic alterations and instability via chromosomal missegregration and initiation of breakage-fusion-bridge cycles. Here, I discuss the role of RNF8 at natural chromosome ends and its (potential) consequences.
- Published
- 2012
10. Ink4a and Arf differentially affect cell proliferation and neural stem cell self-renewal in Bmi1-deficient mice
- Author
-
Maarten van Lohuizen, Jacqueline J.L. Jacobs, Sophia W.M. Bruggeman, Yvan Arsenijevic, Petra van der Stoop, Ellen Tanger, Danielle Hulsman, Silvia Marino, Merel E. Valk-Lingbeek, Karin Kieboom, and Carly Leung
- Subjects
Heterozygote ,Lymphoid Tissue ,Cellular differentiation ,macromolecular substances ,Biology ,Research Communications ,Mice ,Cerebellum ,Proto-Oncogene Proteins ,Tumor Suppressor Protein p14ARF ,Genetics ,Animals ,RNA, Messenger ,Progenitor cell ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Cell Proliferation ,Mice, Knockout ,Neurons ,Polycomb Repressive Complex 1 ,Cell growth ,Genes, p16 ,Multipotent Stem Cells ,Nuclear Proteins ,Cell Differentiation ,Neural stem cell ,body regions ,Mice, Inbred C57BL ,Repressor Proteins ,BMI1 ,Multipotent Stem Cell ,Cancer research ,Stem cell ,Cell aging ,Developmental Biology - Abstract
The Polycomb group (PcG) gene Bmi1 promotes cell proliferation and stem cell self-renewal by repressing the Ink4a/Arf locus. We used a genetic approach to investigate whether Ink4a or Arf is more critical for relaying Bmi1 function in lymphoid cells, neural progenitors, and neural stem cells. We show that Arf is a general target of Bmi1, however particularly in neural stem cells, derepression of Ink4a contributes to Bmi1-/- phenotypes. Additionally, we demonstrate haploinsufficient effects for the Ink4a/Arf locus downstream of Bmi1 in vivo. This suggests differential, cell type-specific roles for Ink4a versus Arf in PcG-mediated (stem) cell cycle control.
- Published
- 2005
11. Significant Role for p16INK4a in p53-Independent Telomere-Directed Senescence
- Author
-
Jacqueline J.L. Jacobs and Titia de Lange
- Subjects
Senescence ,Cell cycle checkpoint ,Fluorescent Antibody Technique ,Biology ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,Small hairpin RNA ,chemistry.chemical_compound ,law ,Humans ,Telomeric Repeat Binding Protein 2 ,neoplasms ,Cells, Cultured ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Agricultural and Biological Sciences(all) ,Biochemistry, Genetics and Molecular Biology(all) ,Fibroblasts ,Telomere ,Immunohistochemistry ,Bromodeoxyuridine ,chemistry ,BMI1 ,Cell culture ,Cancer research ,Suppressor ,Tumor Suppressor Protein p53 ,General Agricultural and Biological Sciences ,DNA Damage - Abstract
Telomere attrition in primary human fibroblasts induces replicative senescence accompanied by activation of the p53 and p16(INK4a)/RB tumor suppressor pathways. Although the contribution of p53 and its target, p21, to telomere-driven senescence have been well established, the role of p16(INK4a) is controversial. Attempts to dissect the significance of p16(INK4a) in response to telomere shortening have been hampered by the concomitant induction of p16(INK4a) by cell culture conditions. To circumvent this problem, we studied the role of p16(INK4a) in the cellular response to acute telomere damage induced by a dominant negative allele of TRF2, TRF2(Delta B Delta M). This approach avoids the confounding aspects of culture stress because parallel cultures with and without telomere damage can be compared. Telomere damage generated with TRF2(Delta B Delta M) resulted in induction of p16(INK4a) in the majority of cells as detected by immunohistochemistry. Inhibition of p16(INK4a) with shRNA or overexpression of BMI1 had a significant effect on the telomere damage response in p53-deficient cells. While p53 deficiency alone only partially abrogated the telomere damage-induced cell cycle arrest, combined inhibition of p16(INK4a) and p53 led to nearly complete bypass of telomere-directed senescence. We conclude that p16(INK4a) contributes to the p53-independent response to telomere damage.
- Published
- 2004
- Full Text
- View/download PDF
12. Polycomb repression: from cellular memory to cellular proliferation and cancer
- Author
-
Jacqueline J.L. Jacobs and Maarten van Lohuizen
- Subjects
Cancer Research ,animal structures ,Polycomb-Group Proteins ,Repressor ,macromolecular substances ,Cell fate determination ,Biology ,medicine.disease_cause ,Neoplasms ,Gene expression ,Genetics ,medicine ,Animals ,Drosophila Proteins ,Humans ,Psychological repression ,Cell growth ,fungi ,Hematopoiesis ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,Oncology ,Carcinogenesis ,Cell Division ,Function (biology) ,Transcription Factors - Abstract
The transcriptional repressors of the Polycomb group (PcG), together with the counteracting Trithorax group (TrxG) proteins, establish a form of cellular memory by regulating gene expression in a heritable fashion at the level of chromatin. This cellular memory function is required for a correct cell fate/behavior, which is not only crucial during development for the generation of a correct body plan but also later in life to prevent cellular transformation. Here, we summarize the rapidly accumulating data that implicate several mammalian PcG members in the control of cellular proliferation and tumorigenesis.
- Published
- 2002
13. Cellular memory of transcriptional states by Polycomb-group proteins
- Author
-
Jacqueline J.L. Jacobs and Maarten van Lohuizen
- Subjects
Genetics ,Cell growth ,Cell ,Repressor ,Cell Biology ,Biology ,Chromatin ,Cell biology ,medicine.anatomical_structure ,Transcription (biology) ,Polycomb-group proteins ,medicine ,Gene silencing ,Homeotic gene ,Developmental Biology - Abstract
The Polycomb–group constitutes an important, widely conserved group of transcriptional repressors best known for their function in stably maintaining the inactive expression patterns of key developmental regulators, including homeotic genes. Together with the counteracting trithorax–group proteins, they establish a form of cellular memory by faithfully maintaining transcription states determined early in embryogenesis. Besides being crucial for the correct execution of developmental programs, Polycomb–group mediated silencing also appears to be involved in controlling cell proliferation. Here we discuss several aspects of Pc–G function: target gene specificity and recognition as well as propagation of inactive chromatin states to subsequent cell generations.
- Published
- 1999
14. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus
- Author
-
Maarten van Lohuizen, Jacqueline J.L. Jacobs, Karin Kieboom, Silvia Marino, and Ronald A. DePinho
- Subjects
Senescence ,Repressor ,Biology ,Mice ,Proto-Oncogene Proteins ,Tumor Suppressor Protein p14ARF ,Animals ,Humans ,Genes, Tumor Suppressor ,Neoplastic transformation ,Cells, Cultured ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Mice, Knockout ,Polycomb Repressive Complex 1 ,Regulation of gene expression ,Genetics ,Multidisciplinary ,Oncogene ,Cell Cycle ,Nuclear Proteins ,Proteins ,Oncogenes ,Fibroblasts ,Cell cycle ,Cell biology ,Repressor Proteins ,Gene Expression Regulation ,BMI1 ,Cell aging ,Cell Division - Abstract
The bmi-1 gene was first isolated as an oncogene that cooperates with c-myc in the generation of mouse lymphomas. We subsequently identified Bmi-1 as a transcriptional repressor belonging to the mouse Polycomb group. The Polycomb group comprises an important, conserved set of proteins that are required to maintain stable repression of specific target genes, such as homeobox-cluster genes, during development. In mice, the absence of bmi-1 expression results in neurological defects and severe proliferative defects in lymphoid cells, whereas bmi-1 overexpression induces lymphomas. Here we show that bmi-1-deficient primary mouse embryonic fibroblasts are impaired in progression into the S phase of the cell cycle and undergo premature senescence. In these fibroblasts and in bmi-1-deficient lymphocytes, the expression of the tumour suppressors p16 and p19Arf, which are encoded by ink4a, is raised markedly. Conversely, overexpression of bmi-1 allows fibroblast immortalization, downregulates expression of p16 and p19Arf and, in combination with H-ras, leads to neoplastic transformation. Removal of ink4a dramatically reduces the lymphoid and neurological defects seen in bmi-1-deficient mice, indicating that ink4a is a critical in vivo target for Bmi-1. Our results connect transcriptional repression by Polycomb-group proteins with cell-cycle control and senescence.
- Published
- 1999
15. MPc2 , a new murine homolog of the Drosophila polycomb protein is a member of the mouse polycomb transcriptional repressor complex 1 1Edited by M. Yaniv
- Author
-
Anton Berns, Neal G. Copeland, Maarten van Lohuizen, Nancy A. Jenkins, Jacqueline J.L. Jacobs, David P. E. Satijn, Jan Willem Voncken, Mark J. Alkema, and Arie P. Otte
- Subjects
Genetics ,Candidate gene ,fungi ,Repressor ,macromolecular substances ,Biology ,Fusion protein ,Non-histone protein ,Transcriptional repressor complex ,Structural Biology ,BMI1 ,Homeotic gene ,Molecular Biology ,Gene - Abstract
The evolutionarily conserved polycomb and trithorax-group genes are required to maintain stable expression patterns of homeotic genes and other target genes throughout development. Here, we report the cloning and characterization of a novel mouse polycomb homolog, MPc2, in addition to the previously described M33 polycomb gene. Co-immunoprecipitations and subnuclear co-localization studies show that MPc2 interacts with the mouse polycomb-group oncoprotein Bmi1 and is a new member of the mouse polycomb multiprotein complex. Gal4DB-MPc2 or -M33 fusion proteins mediate a five- to tenfold repression of stably integrated reporter constructs carrying GAL4 binding sites, demonstrating that these proteins are transcriptional repressors. The MPc2 gene is localized on chromosome 11, in close proximity to the classical mouse mutations tail short (Ts) and rabo torcido (Rbt). Ts and Rbt hemizygous mice display anemia and transformations of the axial skeleton reminiscent of phenotypes observed in mice with mutated polycomb or trithorax-group genes, suggesting that MPc2 is a candidate gene for Ts and Rbt.
- Published
- 1997
16. Loss of Telomere Protection: Consequences and Opportunities
- Author
-
Jacqueline J.L. Jacobs
- Subjects
Senescence ,Genome instability ,Cancer Research ,senescence ,DNA damage ,Biology ,medicine.disease_cause ,Bioinformatics ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,cancer ,030304 developmental biology ,0303 health sciences ,therapy ,DNA-damage ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Shelterin ,telomeres ,genome instability ,3. Good health ,Cell biology ,Telomere ,Oncology ,030220 oncology & carcinogenesis ,Eukaryotic chromosome fine structure ,Perspective Article ,Carcinogenesis ,Function (biology) - Abstract
Telomeres are repetitive sequences at the natural ends of linear eukaryotic chromosomes that protect these from recognition as chromosome breaks. Their ability to do so critically depends on the binding of sufficient quantities of functional shelterin, a six-unit protein complex with specific and crucial roles in telomere maintenance and function. Insufficient telomere length, leading to insufficient concentration of shelterin at chromosome ends, or otherwise crippled shelterin function, causes telomere deprotection. While contributing to aging-related pathologies, loss of telomere protection can act as a barrier to tumorigenesis, as dysfunctional telomeres activate DNA-damage-like checkpoint responses that halt cell proliferation or trigger cell death. In addition, dysfunctional telomeres affect cancer development and progression by being a source of genomic instability. Reviewed here are the different approaches that are being undertaken to investigate the mammalian cellular response to telomere dysfunction and its consequences for cancer. Furthermore, it is discussed how current and future knowledge about the mechanisms underlying telomere damage responses might be applied for diagnostic purposes or therapeutic intervention.
- Published
- 2013
- Full Text
- View/download PDF
17. Senescence: back to telomeres
- Author
-
Jacqueline J.L. Jacobs
- Subjects
Senescence ,Genetics ,Programmed cell death ,DNA damage ,Oncogenes ,Cell Biology ,Telomere ,Biology ,Cell biology ,Neoplasms ,Humans ,Molecular Biology ,Cellular Senescence ,Telomere Shortening ,DNA Damage - Abstract
The demonstration that senescence is caused by both telomere erosion and oncogene-induced accumulation of DNA damage at telomeres.
- Published
- 2013
18. DNA-damage response and repair activities at uncapped telomeres depend on RNF8
- Author
-
Jacqueline J.L. Jacobs and Marieke H. Peuscher
- Subjects
Genome instability ,DNA Repair ,DNA repair ,DNA damage ,Ubiquitin-Protein Ligases ,Immunoblotting ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Genomic Instability ,Histones ,chemistry.chemical_compound ,Mice ,Histone H2A ,Animals ,Humans ,Telomeric Repeat Binding Protein 2 ,Phosphorylation ,Cells, Cultured ,In Situ Hybridization, Fluorescence ,Genetics ,Mice, Knockout ,biology ,Tumor Suppressor Proteins ,Intracellular Signaling Peptides and Proteins ,Ubiquitination ,Cell Biology ,Fibroblasts ,Telomere ,Embryo, Mammalian ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Histone ,HEK293 Cells ,chemistry ,biology.protein ,RNA Interference ,Tumor Suppressor p53-Binding Protein 1 ,DNA ,DNA Damage - Abstract
Loss of telomere protection causes natural chromosome ends to become recognized by DNA-damage response and repair proteins. These events result in ligation of chromosome ends with dysfunctional telomeres, thereby causing chromosomal aberrations on cell division. The control of these potentially dangerous events at deprotected chromosome ends with their unique telomeric chromatin configuration is poorly understood. In particular, it is unknown to what extent bulky modification of telomeric chromatin is involved. Here we show that uncapped telomeres accumulate ubiquitylated histone H2A in a manner dependent on the E3 ligase RNF8. The ability of RNF8 to ubiquitylate telomeric chromatin is associated with its capacity to facilitate accumulation of both 53BP1 and phospho-ATM at uncapped telomeres and to promote non-homologous end-joining of deprotected chromosome ends. In line with the detrimental effect of RNF8 on uncapped telomeres, depletion of RNF8, as well as of the E3 ligase RNF168, reduces telomere-induced genome instability. This indicates that, besides suppressing tumorigenesis by mediating repair of DNA double-strand breaks, RNF8 and RNF168 might enhance cancer development by aggravating telomere-induced genome instability.
- Published
- 2011
19. In vitro genetic screen identifies a cooperative role for LPA signaling and c-Myc in cell transformation
- Author
-
Wouter H. Moolenaar, Ellen Tanger, C Stortelers, E. Verhoeven, J P Lambooij, Panthea Taghavi, Jacqueline J.L. Jacobs, and M van Lohuizen
- Subjects
MAPK/ERK pathway ,Cancer Research ,Cell Survival ,medicine.medical_treatment ,Cell ,Genes, myc ,Biology ,medicine.disease_cause ,chemistry.chemical_compound ,Mice ,Neoplasms ,Lysophosphatidic acid ,Genetics ,medicine ,Animals ,Humans ,Genetic Testing ,Receptors, Lysophosphatidic Acid ,Receptor ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,Cyclin-Dependent Kinase Inhibitor p16 ,Cell growth ,Growth factor ,Fibroblasts ,Embryo, Mammalian ,Cell biology ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,chemistry ,Lysophospholipids ,Carcinogenesis ,Cell Division ,Genetic screen - Abstract
c-Myc drives uncontrolled cell proliferation in various human cancers. However, in mouse embryo fibroblasts (MEFs), c-Myc also induces apoptosis by activating the p19Arf tumor suppressor pathway. Tbx2, a transcriptional repressor of p19Arf, can collaborate with c-Myc by suppressing apoptosis. MEFs overexpressing c-Myc and Tbx2 are immortal but not transformed. We have performed an unbiased genetic screen, which identified 12 oncogenes that collaborate with c-Myc and Tbx2 to transform MEFs in vitro. One of them encodes the LPA2 receptor for the lipid growth factor lysophosphatidic acid (LPA). We find that LPA1 and LPA4, but not LPA3, can reproduce the transforming effect of LPA2. Using pharmacological inhibitors, we show that the in vitro cell transformation induced by LPA receptors is dependent on the Gi-linked ERK and PI3K signaling pathways. The transforming ability of LPA1, LPA2 and LPA4 was confirmed by tumor formation assays in vivo and correlated with prolonged ERK1/2 activation in response to LPA. Our results reveal a direct role for LPA receptor signaling in cell transformation and tumorigenesis in conjunction with c-Myc and reduced p19Arf expression.
- Published
- 2008
20. p16INK4a as a second effector of the telomere damage pathway
- Author
-
Titia de Lange and Jacqueline J.L. Jacobs
- Subjects
Senescence ,Cell cycle checkpoint ,DNA damage ,Effector ,Cell Biology ,Biology ,Telomere ,medicine.disease_cause ,Cell biology ,Apoptosis ,Neoplasms ,Cancer cell ,medicine ,Animals ,Humans ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Molecular Biology ,Cyclin-Dependent Kinase Inhibitor p16 ,Developmental Biology - Abstract
Telomere damage resulting from telomere shortening can potentially suppress tumorigenesis by permanently arresting or eliminating incipient cancer cells. Dysfunctional telomeres activate the canonical DNA damage response pathway, resulting in a p53-mediated G(1)/S arrest and senescence or apoptosis. Experimental induction of telomere damage through inhibition of the telomeric protein TRF2 recapitulates aspects of telomere attrition, including a p53-mediated cell cycle arrest. Using this system, we have shown that telomere damage can also elicit a G(1)/S arrest through the RB-regulator p16INK4a, especially in cells lacking p53 function. Here we discuss the significance of p16INK4a as a second effector of the telomere damage response.
- Published
- 2005
21. Control of the replicative life span of human fibroblasts by p16 and the polycomb protein Bmi-1
- Author
-
Yoko Itahana, Judith Campisi, Jacqueline J.L. Jacobs, Maarten van Lohuizen, Koji Itahana, Ying Zou, Goberdhan P. Dimri, Christian Beauséjour, Jose Luis Martinez, and Vimla Band
- Subjects
Senescence ,DNA Replication ,Telomerase ,Polycomb-Group Proteins ,Retinoblastoma Protein ,p14arf ,Proto-Oncogene Proteins ,Ring finger ,medicine ,Humans ,Nuclear protein ,Molecular Biology ,Cell Growth and Development ,Cells, Cultured ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Helix-Turn-Helix Motifs ,Polycomb Repressive Complex 1 ,biology ,Retinoblastoma protein ,Nuclear Proteins ,Cell Biology ,Fibroblasts ,Telomere ,Molecular biology ,Protein Structure, Tertiary ,Oxygen ,Repressor Proteins ,medicine.anatomical_structure ,Mutation ,biology.protein ,Tumor Suppressor Protein p53 ,Cell aging ,Cell Division - Abstract
The polycomb protein Bmi-1 represses the INK4a locus, which encodes the tumor suppressors p16 and p14(ARF). Here we report that Bmi-1 is downregulated when WI-38 human fibroblasts undergo replicative senescence, but not quiescence, and extends replicative life span when overexpressed. Life span extension by Bmi-1 required the pRb, but not p53, tumor suppressor protein. Deletion analysis showed that the RING finger and helix-turn-helix domains of Bmi-1 were required for life span extension and suppression of p16. Furthermore, a RING finger deletion mutant exhibited dominant negative activity, inducing p16 and premature senescence. Interestingly, presenescent cultures of some, but not all, human fibroblasts contained growth-arrested cells expressing high levels of p16 and apparently arrested by a p53- and telomere-independent mechanism. Bmi-1 selectively extended the life span of these cultures. Low O(2) concentrations had no effect on p16 levels or life span extension by Bmi-1 but reduced expression of the p53 target, p21. We propose that some human fibroblast strains are more sensitive to stress-induced senescence and have both p16-dependent and p53/telomere-dependent pathways of senescence. Our data suggest that Bmi-1 extends the replicative life span of human fibroblasts by suppressing the p16-dependent senescence pathway.
- Published
- 2002
22. The T-box repressors TBX2 and TBX3 specifically regulate the tumor suppressor gene p14ARF via a variant T-site in the initiator
- Author
-
Merel Lingbeek, Maarten van Lohuizen, and Jacqueline J.L. Jacobs
- Subjects
Fetal Proteins ,Brachyury ,Tumor suppressor gene ,Molecular Sequence Data ,Repressor ,Codon, Initiator ,Biology ,medicine.disease_cause ,Biochemistry ,Xbra ,Mice ,Tumor Suppressor Protein p14ARF ,medicine ,Animals ,Humans ,Binding site ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,Psychological repression ,Sequence Deletion ,Genetics ,Mutation ,Binding Sites ,Base Sequence ,Genes, p16 ,Cell Biology ,3T3 Cells ,DNA ,Cell Transformation, Neoplastic ,COS Cells ,Transcription Initiation Site ,T-Box Domain Proteins ,Dimerization - Abstract
The murine tumor suppressor p19(ARF) (p14(ARF) in humans) is thought to fulfill an important protective role in preventing primary cells from oncogenic transformation via its action in the p53 pathway. Several disease-implicated regulators of p19(ARF) are known to date, among which are the T-box genes TBX2, which resides on an amplicon in primary breast tumors, and TBX3, which is mutated in the human developmental disorder Ulnar-Mammary syndrome. Here we identify a variant T-site, matching 13 of 20 nucleotides of a consensus T-site, as the essential TBX2/TBX3-binding element in the human p14(ARF) promoter. Mutant analysis indicates that both the consensus T-box and a C-terminal conserved repression domain are essential for p14(ARF) repression. Whereas the core nucleotides required for interaction of the archetypal T-box protein Brachyury with a consensus T-site are conserved in the variant site, additional flanking nucleotides contribute to the specificity of TBX2 binding. This is illustrated by the inability of TBX1A or Xbra to activate via the variant p14(ARF) T-site. Importantly, this suggests a hitherto unsuspected level of specificity associated with T-box factors and corresponding recognition sites in regulating their target genes in vivo.
- Published
- 2002
23. Senescence bypass screen identifies TBX2, which represses Cdkn2a (p19(ARF)) and is amplified in a subset of human breast cancers
- Author
-
M.J. van de Vijver, Petra M. Nederlof, M van Lohuizen, T van Welsem, Jacqueline J.L. Jacobs, Petra Kristel, Merel Lingbeek, P Keblusek, E Y Koh, George Q. Daley, Els Robanus-Maandag, and Other departments
- Subjects
Genes, BRCA1 ,Cell Cycle Proteins ,medicine.disease_cause ,Mice ,CDKN2A ,Tumor Suppressor Protein p14ARF ,Tumor Cells, Cultured ,E2F1 ,Promoter Regions, Genetic ,Cells, Cultured ,Cellular Senescence ,Regulation of gene expression ,Polycomb Repressive Complex 1 ,Nuclear Proteins ,Neoplasm Proteins ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,COS Cells ,Female ,Transcription Factor DP1 ,Tumor suppressor gene ,Breast Neoplasms ,Biology ,Adenocarcinoma ,Transfection ,Proto-Oncogene Proteins c-myc ,Proto-Oncogene Proteins p21(ras) ,Neoplastic Syndromes, Hereditary ,Proto-Oncogene Proteins ,Genetics ,medicine ,Animals ,Humans ,Cyclin-Dependent Kinase Inhibitor p16 ,Oncogene ,Gene Amplification ,Proteins ,Oncogenes ,Fibroblasts ,E2F Transcription Factors ,Repressor Proteins ,BMI1 ,Protein Biosynthesis ,Cancer research ,Carcinogenesis ,Carrier Proteins ,T-Box Domain Proteins ,E2F1 Transcription Factor ,Gene Deletion ,Genetic screen ,Chromosomes, Human, Pair 17 ,Retinoblastoma-Binding Protein 1 ,Transcription Factors - Abstract
To identify new immortalizing genes with potential roles in tumorigenesis, we performed a genetic screen aimed to bypass the rapid and tight senescence arrest of primary fibroblasts deficient for the oncogene Bmi1. We identified the T-box member TBX2 as a potent immortalizing gene that acts by downregulating Cdkn2a (p19(ARF)). TBX2 represses the Cdkn2a (p19(ARF)) promoter and attenuates E2F1, Myc or HRAS-mediated induction of Cdkn2a (p19(ARF)). We found TBX2 to be amplified in a subset of primary human breast cancers, indicating that it might contribute to breast cancer development.
- Published
- 2000
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.