64 results on '"Motoki Takaku"'
Search Results
2. GATA3 Truncation Mutants Alter EMT Related Gene Expression via Partial Motif Recognition in Luminal Breast Cancer Cells
- Author
-
Mika Saotome, Deepak Balakrishnan Poduval, Renju Nair, Mikhala Cooper, and Motoki Takaku
- Subjects
breast cancer ,GATA3 ,GATA-3 ,somatic mutations ,luminal breast cancer ,epithelial-to-mesenchymal transition ,Genetics ,QH426-470 - Abstract
GATA3 is known to be one of the most frequently mutated genes in breast cancer. More than 10% of breast tumors carry mutations in this gene. However, the functional consequence of GATA3 mutations is still largely unknown. Clinical data suggest that different types of GATA3 mutations may have distinct roles in breast cancer characterization. In this study, we have established three luminal breast cancer cell lines that stably express different truncation mutants (X308 splice site deletion, C321 frameshift, and A333 frameshift mutants) found in breast cancer patients. Transcriptome analysis identified common and distinct gene expression patterns in these GATA3 mutant cell lines. In particular, the impacts on epithelial-to-mesenchymal transition (EMT) related genes are similar across these mutant cell lines. Chromatin localization of the mutants is highly overlapped and exhibits non-canonical motif enrichment. Interestingly, the A333 frameshift mutant expressed cells displayed the most significant impact on the GATA3 binding compared to X308 splice site deletion and C321fs mutants expressed cells. Our results suggest the common and different roles of GATA3 truncation mutations during luminal breast cancer development.
- Published
- 2022
- Full Text
- View/download PDF
3. Author Correction: CHD3 helicase domain mutations cause a neurodevelopmental syndrome with macrocephaly and impaired speech and language
- Author
-
Lot Snijders Blok, Justine Rousseau, Joanna Twist, Sophie Ehresmann, Motoki Takaku, Hanka Venselaar, Lance H. Rodan, Catherine B. Nowak, Jessica Douglas, Kathryn J. Swoboda, Marcie A. Steeves, Inderneel Sahai, Connie T. R. M. Stumpel, Alexander P. A. Stegmann, Patricia Wheeler, Marcia Willing, Elise Fiala, Aaina Kochhar, William T. Gibson, Ana S. A. Cohen, Ruky Agbahovbe, A. Micheil Innes, P. Y. Billie Au, Julia Rankin, Ilse J. Anderson, Steven A. Skinner, Raymond J. Louie, Hannah E. Warren, Alexandra Afenjar, Boris Keren, Caroline Nava, Julien Buratti, Arnaud Isapof, Diana Rodriguez, Raymond Lewandowski, Jennifer Propst, Ton van Essen, Murim Choi, Sangmoon Lee, Jong H. Chae, Susan Price, Rhonda E. Schnur, Ganka Douglas, Ingrid M. Wentzensen, Christiane Zweier, André Reis, Martin G. Bialer, Christine Moore, Marije Koopmans, Eva H. Brilstra, Glen R. Monroe, Koen L. I. van Gassen, Ellen van Binsbergen, Ruth Newbury-Ecob, Lucy Bownass, Ingrid Bader, Johannes A. Mayr, Saskia B. Wortmann, Kathy J. Jakielski, Edythe A. Strand, Katja Kloth, Tatjana Bierhals, The DDD study, John D. Roberts, Robert M. Petrovich, Shinichi Machida, Hitoshi Kurumizaka, Stefan Lelieveld, Rolph Pfundt, Sandra Jansen, Pelagia Deriziotis, Laurence Faivre, Julien Thevenon, Mirna Assoum, Lawrence Shriberg, Tjitske Kleefstra, Han G. Brunner, Paul A. Wade, Simon E. Fisher, and Philippe M. Campeau
- Subjects
Science - Abstract
The HTML and PDF versions of this Article were updated after publication to remove images of one individual from Figure 1.
- Published
- 2019
- Full Text
- View/download PDF
4. DNA methylation in mice is influenced by genetics as well as sex and life experience
- Author
-
Sara A. Grimm, Takashi Shimbo, Motoki Takaku, James W. Thomas, Scott Auerbach, Brian D. Bennett, John R. Bucher, Adam B. Burkholder, Frank Day, Ying Du, Christopher G. Duncan, John E. French, Julie F. Foley, Jianying Li, B. Alex Merrick, Raymond R. Tice, Tianyuan Wang, Xiaojiang Xu, NISC Comparative Sequencing Program, Pierre R. Bushel, David C. Fargo, James C. Mullikin, and Paul A. Wade
- Subjects
Science - Abstract
DNA methylation is an epigenetic mark involved in gene regulation. Here the authors investigate the extent to which genetics, sex and pregnancy influence genomic DNA methylation in mice, providing evidence of the stability of CpG methylation across generation and suggest that CpG methylation may serve as an epigenetic record of life events in somatic tissues at loci whose expression is linked to the relevant biology.
- Published
- 2019
- Full Text
- View/download PDF
5. CHD3 helicase domain mutations cause a neurodevelopmental syndrome with macrocephaly and impaired speech and language
- Author
-
Lot Snijders Blok, Justine Rousseau, Joanna Twist, Sophie Ehresmann, Motoki Takaku, Hanka Venselaar, Lance H. Rodan, Catherine B. Nowak, Jessica Douglas, Kathryn J. Swoboda, Marcie A. Steeves, Inderneel Sahai, Connie T. R. M. Stumpel, Alexander P. A. Stegmann, Patricia Wheeler, Marcia Willing, Elise Fiala, Aaina Kochhar, William T. Gibson, Ana S. A. Cohen, Ruky Agbahovbe, A. Micheil Innes, P. Y. Billie Au, Julia Rankin, Ilse J. Anderson, Steven A. Skinner, Raymond J. Louie, Hannah E. Warren, Alexandra Afenjar, Boris Keren, Caroline Nava, Julien Buratti, Arnaud Isapof, Diana Rodriguez, Raymond Lewandowski, Jennifer Propst, Ton van Essen, Murim Choi, Sangmoon Lee, Jong H. Chae, Susan Price, Rhonda E. Schnur, Ganka Douglas, Ingrid M. Wentzensen, Christiane Zweier, André Reis, Martin G. Bialer, Christine Moore, Marije Koopmans, Eva H. Brilstra, Glen R. Monroe, Koen L. I. van Gassen, Ellen van Binsbergen, Ruth Newbury-Ecob, Lucy Bownass, Ingrid Bader, Johannes A. Mayr, Saskia B. Wortmann, Kathy J. Jakielski, Edythe A. Strand, Katja Kloth, Tatjana Bierhals, The DDD study, John D. Roberts, Robert M. Petrovich, Shinichi Machida, Hitoshi Kurumizaka, Stefan Lelieveld, Rolph Pfundt, Sandra Jansen, Pelagia Deriziotis, Laurence Faivre, Julien Thevenon, Mirna Assoum, Lawrence Shriberg, Tjitske Kleefstra, Han G. Brunner, Paul A. Wade, Simon E. Fisher, and Philippe M. Campeau
- Subjects
Science - Abstract
Chromodomain Helicase DNA-binding (CHD) proteins have been implicated in neurodevelopmental processes. Here, the authors identify missense variants in CHD3 that disturb its chromatin remodeling activities and cause a neurodevelopmental disorder with macrocephaly and speech and language impairment.
- Published
- 2018
- Full Text
- View/download PDF
6. GATA3 zinc finger 2 mutations reprogram the breast cancer transcriptional network
- Author
-
Motoki Takaku, Sara A. Grimm, John D. Roberts, Kaliopi Chrysovergis, Brian D. Bennett, Page Myers, Lalith Perera, Charles J. Tucker, Charles M. Perou, and Paul A. Wade
- Subjects
Science - Abstract
In breast cancer GATA3 is known to be frequently mutated, but the function of these mutations is unclear. Here, the authors utilise CRISPR-Cas9 to model frame-shift mutations in zinc finger 2 of GATA3, highlighting that GATA3 mutation can have gain- or loss-of function effects in breast cancer.
- Published
- 2018
- Full Text
- View/download PDF
7. High-quality ChIP-seq analysis of MBD3 in human breast cancer cells
- Author
-
Takashi Shimbo, Motoki Takaku, and Paul A. Wade
- Subjects
Genetics ,QH426-470 - Abstract
Chromatin accessibility is tightly regulated by multiple factors/mechanisms to establish different cell type-specific gene expression programs from a single genome. Dysregulation of this process can lead to diseases including cancer. The Mi-2/nucleosome remodeling and deacetylase (NuRD) complex is thought to orchestrate chromatin structure using its intrinsic nucleosome remodeling and histone deacetylase activities. However, the detailed mechanisms by which the NuRD complex regulates chromatin structure in vivo are not yet known. To explore the regulatory mechanisms of the NuRD complex, we mapped genome-wide localization of MBD3, a structural component of NuRD, in a human breast cancer cell line (MDA-MB-231) using a modified ChIP-seq protocol. Our data showed high quality localization information (i.e., high mapping efficiency and low PCR duplication rate) and excellent consistency between biological replicates. The data are deposited in the Gene Expression Omnibus (GSE76116). Keywords: ChIP-seq, Mi-2/NuRD, Breast cancer
- Published
- 2016
- Full Text
- View/download PDF
8. Supplementary Table from Autocrine GMCSF Signaling Contributes to Growth of HER2+ Breast Leptomeningeal Carcinomatosis
- Author
-
Rahul Jandial, Motoki Takaku, Clara Chen, Bony De Kumar, Antariksh Tyagi, Mika Saotome, Arunoday Bhan, and Khairul I. Ansari
- Abstract
Supplementary Table from Autocrine GMCSF Signaling Contributes to Growth of HER2+ Breast Leptomeningeal Carcinomatosis
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Autocrine GMCSF Signaling Contributes to Growth of HER2+ Breast Leptomeningeal Carcinomatosis
- Author
-
Rahul Jandial, Motoki Takaku, Clara Chen, Bony De Kumar, Antariksh Tyagi, Mika Saotome, Arunoday Bhan, and Khairul I. Ansari
- Abstract
Leptomeningeal carcinomatosis (LC) occurs when tumor cells spread to the cerebrospinal fluid–containing leptomeninges surrounding the brain and spinal cord. LC is an ominous complication of cancer with a dire prognosis. Although any malignancy can spread to the leptomeninges, breast cancer, particularly the HER2+ subtype, is its most common origin. HER2+ breast LC (HER2+ LC) remains incurable, with few treatment options, and the molecular mechanisms underlying proliferation of HER2+ breast cancer cells in the acellular, protein, and cytokine-poor leptomeningeal environment remain elusive. Therefore, we sought to characterize signaling pathways that drive HER2+ LC development as well as those that restrict its growth to leptomeninges. Primary HER2+ LC patient-derived (“Lepto”) cell lines in coculture with various central nervous system (CNS) cell types revealed that oligodendrocyte progenitor cells (OPC), the largest population of dividing cells in the CNS, inhibited HER2+ LC growth in vitro and in vivo, thereby limiting the spread of HER2+ LC beyond the leptomeninges. Cytokine array–based analyses identified Lepto cell–secreted GMCSF as an oncogenic autocrine driver of HER2+ LC growth. LC/MS-MS-based analyses revealed that the OPC-derived protein TPP1 proteolytically degrades GMCSF, decreasing GMCSF signaling and leading to suppression of HER2+ LC growth and limiting its spread. Finally, intrathecal delivery of neutralizing anti-GMCSF antibodies and a pan-Aurora kinase inhibitor (CCT137690) synergistically inhibited GMCSF and suppressed activity of GMCSF effectors, reducing HER2+ LC growth in vivo. Thus, OPC suppress GMCSF-driven growth of HER2+ LC in the leptomeningeal environment, providing a potential targetable axis.Significance:This study characterizes molecular mechanisms that drive HER2+ leptomeningeal carcinomatosis and demonstrates the efficacy of anti-GMCSF antibodies and pan-Aurora kinase inhibitors against this disease.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure from Autocrine GMCSF Signaling Contributes to Growth of HER2+ Breast Leptomeningeal Carcinomatosis
- Author
-
Rahul Jandial, Motoki Takaku, Clara Chen, Bony De Kumar, Antariksh Tyagi, Mika Saotome, Arunoday Bhan, and Khairul I. Ansari
- Abstract
Supplementary Figure from Autocrine GMCSF Signaling Contributes to Growth of HER2+ Breast Leptomeningeal Carcinomatosis
- Published
- 2023
- Full Text
- View/download PDF
11. Genomic transcription factor binding site selection is edited by the chromatin remodeling factor CHD4
- Author
-
Motoki Takaku, Mika Saotome, Deepak Poduval, Sara Grimm, Aerica Nagornyuk, Sakuntha Gunarathna, Takashi Shimbo, and Paul Wade
- Abstract
Biologically precise enhancer licensing by lineage-determining transcription factors enables activation of transcripts appropriate to biological demand and prevents deleterious gene activation. This essential process is challenged by the millions of matches to most transcription factor binding motifs present in many eukaryotic genomes, leading to questions about how transcription factors achieve the exquisite specificity required. The importance of chromatin remodeling factors to enhancer activation is highlighted by their frequent mutation in developmental disorders and in cancer. Here we determine the roles of CHD4 to enhancer licensing and maintenance in breast cancer cells and during cellular reprogramming. In unchallenged basal breast cancer cells, CHD4 modulates chromatin accessibility at transcription factor binding sites; its depletion leads to altered motif scanning and redistribution of transcription factors to sites not previously occupied. During GATA3-mediated cellular reprogramming, CHD4 activity is necessary to prevent inappropriate chromatin opening and enhancer licensing. Mechanistically, CHD4 competes with transcription factor-DNA interaction by promoting nucleosome positioning over binding motifs. We propose that CHD4 acts as a chromatin proof-reading enzyme that prevents inappropriate gene expression by editing binding site selection by transcription factors.
- Published
- 2023
- Full Text
- View/download PDF
12. Abstract P3-09-12: Breast cancer derived GATA3 mutations disrupt luminal transcriptional network
- Author
-
Motoki Takaku, Mika Saotome, and Renju Nair
- Subjects
Cancer Research ,Oncology - Abstract
GATA3 has been identified as one of the most frequently mutated genes in breast cancer. In the METABRIC cohort, among 1,980 patient cases, 230 breast cancers harbored GATA3 mutations (~12%). 75% of the GATA mutations were observed in luminal breast tumors (47% in luminal A, 28% in luminal B). The recent genomic data in metastatic breast cancer also showed that the frequency of GATA3 somatic mutations was even higher in the metastatic breast cancer cohort. Lung, lymph nodes, and brain metastases were observed in the GATA3 mutant breast cancer patients. Based on these patient genomic data, GATA3 mutations have been considered as cancer drivers, yet the functional consequences of GATA3 mutations are underexplored. We and other groups previously identified that patients carrying GATA3 mutations have diverse clinical features. More than 70% of cases are small nucleotide deletions or insertions (indel), while less than 30% are missense mutations. By classifying the GATA3 indel mutations into 4 groups, we observed distinct clinical features. Somatic mutations found in the GATA3 second zinc-finger domain (ZnFn2) are significantly associated with poorer patient outcomes including worse patient survival. ZnFn2 mutations are predominantly found in luminal B breast tumors, while splice site mutations are frequently found in luminal A breast tumors and associated with better patient survival. These distinct clinical features clearly suggest the differential impacts of GATA3 mutations on breast cancer cells. To dissect the function of GATA3 mutants, we developed a GATA3 mutant cell line (R330fs/WT T47D cells) by CRISPR, which endogenously expresses a heterozygous R330 frame-shift (R330fs) mutant. R330fs mutations are found in multiple data cohorts. The majority (>90%) of GATA3 mutations are heterozygous. Therefore, this R330fs mutant T47D cell line mimics the type of alteration found in patients. Importantly, we found that the R330fs mutation induces transcriptional reprogramming in T47D luminal breast cancer cells leading to more aggressive phenotypes such as faster tumor growth. In the mutant cells, many epithelial marker genes (such as progesterone receptor and TFF1) were down-regulated while mesenchymal marker genes (such as TWIST1 and SNAI2) are up-regulated suggesting that R330fs induces a partial EMT in T47D cells. We also found that transcriptional changes in the R330fs mutant cells were strongly associated with redistribution of GATA3, Estrogen Receptor alpha, and FOXA1. Gene expression profiles in the other GATA3 mutant breast cancer cell lines also suggest similar genomic alterations. These results suggest the active roles of GATA3 mutations during breast cancer development. Citation Format: Motoki Takaku, Mika Saotome, Renju Nair. Breast cancer derived GATA3 mutations disrupt luminal transcriptional network [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P3-09-12.
- Published
- 2022
- Full Text
- View/download PDF
13. P4HTM: A Novel Downstream Target of GATA3 in Breast Cancer
- Author
-
Sarah C. DiDonna, Aerica Nagornyuk, Neeta Adhikari, Mamoru Takada, and Motoki Takaku
- Abstract
Breast cancer continues to be a major cause of death among women. The GATA3 gene is often overexpressed in breast cancer and is widely used to support a diagnosis. However, lower expression of GATA3 has been linked to poorer prognosis along with frequent gene mutations. Therefore, the role of GATA3 in breast cancer appears to be context specific. This study aims to identify a new downstream target of GATA3 to better understand its regulatory network. Clinical data analysis identified the prolyl 4-hydroxylase transmembrane protein (P4HTM) as one of the most highly co-expressed genes with GATA3. Immunohistochemical staining of breast tumors confirms co-expression between GATA3 and P4HTM at the protein level. Similar to GATA3, P4HTM expression levels are linked to patient prognosis, with lower levels indicating poorer survival. Genomics data found that GATA3 binds to the P4HTM locus, and that ectopic expression of GATA3 in basal breast cancer cells increases the P4HTM transcript level. These results collectively suggest that P4HTM is a novel downstream target of GATA3 in breast cancer and is involved in tumor progression.
- Published
- 2023
- Full Text
- View/download PDF
14. Autocrine GMCSF Signaling Contributes to Growth of HER2+ Breast Leptomeningeal Carcinomatosis
- Author
-
Clara Chen, Mika Saotome, Motoki Takaku, Rahul Jandial, Antariksh Tyagi, Arunoday Bhan, Bony De Kumar, and Khairul I. Ansari
- Subjects
Cancer Research ,Cell type ,education.field_of_study ,Chemistry ,medicine.medical_treatment ,Leptomeninges ,Population ,Cancer ,medicine.disease ,Cytokine ,Oncology ,Cell culture ,medicine ,Cancer research ,Signal transduction ,skin and connective tissue diseases ,Autocrine signalling ,education - Abstract
Leptomeningeal carcinomatosis (LC) occurs when tumor cells spread to the cerebrospinal fluid–containing leptomeninges surrounding the brain and spinal cord. LC is an ominous complication of cancer with a dire prognosis. Although any malignancy can spread to the leptomeninges, breast cancer, particularly the HER2+ subtype, is its most common origin. HER2+ breast LC (HER2+ LC) remains incurable, with few treatment options, and the molecular mechanisms underlying proliferation of HER2+ breast cancer cells in the acellular, protein, and cytokine-poor leptomeningeal environment remain elusive. Therefore, we sought to characterize signaling pathways that drive HER2+ LC development as well as those that restrict its growth to leptomeninges. Primary HER2+ LC patient-derived (“Lepto”) cell lines in coculture with various central nervous system (CNS) cell types revealed that oligodendrocyte progenitor cells (OPC), the largest population of dividing cells in the CNS, inhibited HER2+ LC growth in vitro and in vivo, thereby limiting the spread of HER2+ LC beyond the leptomeninges. Cytokine array–based analyses identified Lepto cell–secreted GMCSF as an oncogenic autocrine driver of HER2+ LC growth. LC/MS-MS-based analyses revealed that the OPC-derived protein TPP1 proteolytically degrades GMCSF, decreasing GMCSF signaling and leading to suppression of HER2+ LC growth and limiting its spread. Finally, intrathecal delivery of neutralizing anti-GMCSF antibodies and a pan-Aurora kinase inhibitor (CCT137690) synergistically inhibited GMCSF and suppressed activity of GMCSF effectors, reducing HER2+ LC growth in vivo. Thus, OPC suppress GMCSF-driven growth of HER2+ LC in the leptomeningeal environment, providing a potential targetable axis. Significance: This study characterizes molecular mechanisms that drive HER2+ leptomeningeal carcinomatosis and demonstrates the efficacy of anti-GMCSF antibodies and pan-Aurora kinase inhibitors against this disease.
- Published
- 2021
- Full Text
- View/download PDF
15. ATAC-Seq Optimization for Cancer Epigenetics Research
- Author
-
Motoki Takaku, Archana Dhasarathy, Atrayee Bhattacharya, Atrayee Ray, and Mikhala Cooper
- Subjects
General Immunology and Microbiology ,Neoplasms ,General Chemical Engineering ,General Neuroscience ,Chromatin Immunoprecipitation Sequencing ,High-Throughput Nucleotide Sequencing ,DNA ,Sequence Analysis, DNA ,Chromatin ,Article ,General Biochemistry, Genetics and Molecular Biology ,Epigenesis, Genetic ,Transcription Factors - Abstract
The assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) probes deoxyribonucleic acid (DNA) accessibility using the hyperactive Tn5 transposase. Tn5 cuts and ligates adapters for high-throughput sequencing within accessible chromatin regions. In eukaryotic cells, genomic DNA is packaged into chromatin, a complex of DNA, histones, and other proteins, which acts as a physical barrier to the transcriptional machinery. In response to extrinsic signals, transcription factors recruit chromatin remodeling complexes to enable access to the transcriptional machinery for gene activation. Therefore, identifying open chromatin regions is useful when monitoring enhancer and gene promoter activities during biological events such as cancer progression. Since this protocol is easy to use and has a low cell input requirement, ATAC-seq has been widely adopted to define open chromatin regions in various cell types, including cancer cells. For successful data acquisition, several parameters need to be considered when preparing ATAC-seq libraries. Among them, the choice of cell lysis buffer, the titration of the Tn5 enzyme, and the starting volume of cells are crucial for ATAC-seq library preparation in cancer cells. Optimization is essential for generating high-quality data. Here, we provide a detailed description of the ATAC-seq optimization methods for epithelial cell types.
- Published
- 2022
- Full Text
- View/download PDF
16. GATA3 Truncation Mutants Alter EMT Related Gene Expression
- Author
-
Mika, Saotome, Deepak Balakrishnan, Poduval, Renju, Nair, Mikhala, Cooper, and Motoki, Takaku
- Abstract
GATA3 is known to be one of the most frequently mutated genes in breast cancer. More than 10% of breast tumors carry mutations in this gene. However, the functional consequence of GATA3 mutations is still largely unknown. Clinical data suggest that different types of GATA3 mutations may have distinct roles in breast cancer characterization. In this study, we have established three luminal breast cancer cell lines that stably express different truncation mutants (X308 splice site deletion, C321 frameshift, and A333 frameshift mutants) found in breast cancer patients. Transcriptome analysis identified common and distinct gene expression patterns in these GATA3 mutant cell lines. In particular, the impacts on epithelial-to-mesenchymal transition (EMT) related genes are similar across these mutant cell lines. Chromatin localization of the mutants is highly overlapped and exhibits non-canonical motif enrichment. Interestingly, the A333 frameshift mutant expressed cells displayed the most significant impact on the GATA3 binding compared to X308 splice site deletion and C321fs mutants expressed cells. Our results suggest the common and different roles of GATA3 truncation mutations during luminal breast cancer development.
- Published
- 2021
17. Autocrine GMCSF Signaling Contributes to Growth of HER2
- Author
-
Khairul I, Ansari, Arunoday, Bhan, Mika, Saotome, Antariksh, Tyagi, Bony, De Kumar, Clara, Chen, Motoki, Takaku, and Rahul, Jandial
- Subjects
Oncogene Proteins ,Cell Survival ,Receptor, ErbB-2 ,Gene Expression ,Granulocyte-Macrophage Colony-Stimulating Factor ,Breast Neoplasms ,Xenograft Model Antitumor Assays ,Article ,Autocrine Communication ,Disease Models, Animal ,Mice ,Cell Line, Tumor ,Animals ,Humans ,skin and connective tissue diseases ,Meningeal Carcinomatosis ,Protein Kinase Inhibitors ,Cell Proliferation ,Signal Transduction - Abstract
Leptomeningeal carcinomatosis (LC) occurs when tumor cells spread to the cerebrospinal fluid-containing leptomeninges surrounding the brain and spinal cord. LC is an ominous complication of cancer with a dire prognosis. Although any malignancy can spread to the leptomeninges, breast cancer, particularly the HER2+ subtype, is its most common origin. HER2+ breast LC (HER2+ LC) remains incurable, with few treatment options, and the molecular mechanisms underlying proliferation of HER2+ breast cancer cells in the acellular, protein, and cytokine-poor leptomeningeal environment remain elusive. Therefore, we sought to characterize signaling pathways that drive HER2+ LC development as well as those that restrict its growth to leptomeninges. Primary HER2+ LC patient-derived (“Lepto”) cell lines in co-culture with various central nervous system (CNS) cell types revealed that oligodendrocyte progenitor cells (OPC), the largest population of dividing cells in the CNS, inhibited HER2+ LC growth in vitro and in vivo, thereby limiting the spread of HER2+ LC beyond the leptomeninges. Cytokine array-based analyses identified Lepto cell-secreted granulocyte-macrophage colony-stimulating factor (GM-CSF) as an oncogenic autocrine driver of HER2+ LC growth. Liquid chromatography-tandem mass spectrometry-based analyses revealed that the OPC-derived protein TPP1 proteolytically degrades GM-CSF, decreasing GM-CSF signaling and leading to suppression of HER2+ LC growth and limiting its spread. Lastly, intrathecal delivery of neutralizing anti-GM-CSF antibodies and a pan-Aurora kinase inhibitor (CCT137690) synergistically inhibited GM-CSF and suppressed activity of GM-CSF effectors, reducing HER2+ LC growth in vivo. Thus, OPC suppress GM-CSF-driven growth of HER2+ LC in the leptomeningeal environment, providing a potential targetable axis.
- Published
- 2021
18. Cancer-specific mutation of GATA3 disrupts the transcriptional regulatory network governed by Estrogen Receptor alpha, FOXA1 and GATA3
- Author
-
Motoki Takaku, Paul A. Wade, Bony De Kumar, Brian D. Bennett, and Sara A. Grimm
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Transcription, Genetic ,Mutant ,Estrogen receptor ,Breast Neoplasms ,GATA3 Transcription Factor ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Gene Regulatory Networks ,Transcription factor ,030304 developmental biology ,0303 health sciences ,Gene regulation, Chromatin and Epigenetics ,GATA3 ,Estrogen Receptor alpha ,Cancer ,medicine.disease ,Chromatin ,Cell biology ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Mutation ,Female ,FOXA1 ,Estrogen receptor alpha - Abstract
Estrogen receptors (ER) are activated by the steroid hormone 17β-estradiol. Estrogen receptor alpha (ER-α) forms a regulatory network in mammary epithelial cells and in breast cancer with the transcription factors FOXA1 and GATA3. GATA3 is one of the most frequently mutated genes in breast cancer and is capable of specifying chromatin localization of FOXA1 and ER-α. How GATA3 mutations found in breast cancer impact genomic localization of ER-α and the transcriptional network downstream of ER-α and FOXA1 remains unclear. Here, we investigate the function of a recurrent patient-derived GATA3 mutation (R330fs) on this regulatory network. Genomic analysis indicates that the R330fs mutant can disrupt localization of ER-α and FOXA1. Loci co-bound by all three factors are enriched for genes integral to mammary gland development as well as epithelial cell biology. This gene set is differentially regulated in GATA3 mutant cells in culture and in tumors bearing similar mutations in vivo. The altered distribution of ER-α and FOXA1 in GATA3-mutant cells is associated with altered chromatin architecture, which leads to differential gene expression. These results suggest an active role for GATA3 zinc finger 2 mutants in ER-α positive breast tumors.
- Published
- 2020
19. Reversing SKI–SMAD4-mediated suppression is essential for TH17 cell differentiation
- Author
-
Donald N. Cook, Jonathan S. Serody, Ge Zhang, Jenny P.-Y. Ting, Xian Chen, Yisong Y. Wan, Xiaojiang Xu, Paul A. Wade, Bing Wu, Song Zhang, Qing Kong, Seddon Y. Thomas, Motoki Takaku, Wei-Chun Chou, Liyun Zou, and Ai Di Gu
- Subjects
0301 basic medicine ,Orphan receptor ,animal structures ,Multidisciplinary ,Histone acetylation and deacetylation ,Cellular differentiation ,Biology ,Molecular biology ,digestive system diseases ,Cell biology ,03 medical and health sciences ,Retinoic acid receptor ,chemistry.chemical_compound ,030104 developmental biology ,chemistry ,RAR-related orphan receptor gamma ,embryonic structures ,Signal transduction ,Receptor ,Transforming growth factor - Abstract
T helper 17 (TH17) cells are critically involved in host defence, inflammation, and autoimmunity. Transforming growth factor β (TGFβ) is instrumental in TH17 cell differentiation by cooperating with interleukin-6 (refs 6, 7). Yet, the mechanism by which TGFβ enables TH17 cell differentiation remains elusive. Here we reveal that TGFβ enables TH17 cell differentiation by reversing SKI-SMAD4-mediated suppression of the expression of the retinoic acid receptor (RAR)-related orphan receptor γt (RORγt). We found that, unlike wild-type T cells, SMAD4-deficient T cells differentiate into TH17 cells in the absence of TGFβ signalling in a RORγt-dependent manner. Ectopic SMAD4 expression suppresses RORγt expression and TH17 cell differentiation of SMAD4-deficient T cells. However, TGFβ neutralizes SMAD4-mediated suppression without affecting SMAD4 binding to the Rorc locus. Proteomic analysis revealed that SMAD4 interacts with SKI, a transcriptional repressor that is degraded upon TGFβ stimulation. SKI controls histone acetylation and deacetylation of the Rorc locus and TH17 cell differentiation via SMAD4: ectopic SKI expression inhibits H3K9 acetylation of the Rorc locus, Rorc expression, and TH17 cell differentiation in a SMAD4-dependent manner. Therefore, TGFβ-induced disruption of SKI reverses SKI-SMAD4-mediated suppression of RORγt to enable TH17 cell differentiation. This study reveals a critical mechanism by which TGFβ controls TH17 cell differentiation and uncovers the SKI-SMAD4 axis as a potential therapeutic target for treating TH17-related diseases.
- Published
- 2017
- Full Text
- View/download PDF
20. MyD88-dependent Dendritic and Epithelial Cell Crosstalk Orchestrates Immune Responses to Allergens
- Author
-
Keiko Nakano, Miranda R. Lyons-Cohen, Seddon Y. Thomas, G S Whitehead, Hideki Nakano, Donald N. Cook, Paul A. Wade, Motoki Takaku, Kymberly M. Gowdy, Xiaojiang Xu, and James M. Ward
- Subjects
0301 basic medicine ,Cell type ,Cell signaling ,Cellular differentiation ,Immunology ,Cell Communication ,Respiratory Mucosa ,Biology ,Article ,Allergic inflammation ,Allergic sensitization ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Administration, Inhalation ,Hypersensitivity ,Immunology and Allergy ,Animals ,Humans ,Regulation of gene expression ,Mice, Knockout ,hemic and immune systems ,Cell Differentiation ,Dendritic Cells ,respiratory system ,Allergens ,Asthma ,3. Good health ,Cell biology ,respiratory tract diseases ,Eosinophils ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Gene Expression Regulation ,Myeloid Differentiation Factor 88 ,Th17 Cells ,Immunization ,Signal transduction ,030215 immunology ,Signal Transduction - Abstract
Sensitization to inhaled allergens is dependent on activation of conventional dendritic cells (cDCs) and on the adaptor molecule, MyD88. However, many cell types in the lung express Myd88, and it is unclear how signaling in these different cell types reprograms cDCs and leads to allergic inflammation of the airway. By combining ATAC-seq with RNA profiling, we found that MyD88 signaling in cDCs maintained open chromatin at select loci even at steady state, allowing genes to be rapidly induced during allergic sensitization. A distinct set of genes related to metabolism was indirectly controlled in cDCs through MyD88 signaling in airway epithelial cells (ECs). In mouse models of asthma, Myd88 expression in ECs was critical for eosinophilic inflammation, whereas Myd88 expression in cDCs was required for Th17 cell differentiation and consequent airway neutrophilia. Thus, both cell-intrinsic and cell-extrinsic MyD88 signaling controls gene expression in cDCs and orchestrates immune responses to inhaled allergens.
- Published
- 2017
21. The CHD4-related syndrome: a comprehensive investigation of the clinical spectrum, genotype-phenotype correlations, and molecular basis
- Author
-
Marie Falkenberg Smeland, Alina Kurolap, Michael J. Gambello, Ariel F. Martinez, Livija Medne, Melita Irving, Elizabeth Roeder, Holly Dubbs, Robert M. Petrovich, Elaine H. Zackai, Motoki Takaku, Kimberly Nugent, Bruce D. Gelb, Peter D. Turnpenny, Michael Parker, Maximilian Muenke, Thomas Smol, Arie van Haeringen, Hanne Hove, Hitoshi Kurumizaka, Sandra Whalen, Boris Keren, Philippe M. Campeau, Samantha A. Schrier Vergano, Lior Cohen, Pauline Terhal, Amy Kenney, Paul A. Wade, Jill Clayton-Smith, Jamal Ghoumid, Shane C. Quinonez, John Roberts, Katherine Lachlan, Mahim Jain, Estelle Colin, Melissa Rumple, Solveig Heide, Kay Metcalfe, Alban Ziegler, Hayley P. Lazar, Elizabeth T. DeChene, Cara M. Skraban, Michael Wright, Karin Weiss, Danielle Monteil, Tamar Paperna, Avni Santani, Hagit Baris Feldman, Bryan L. Krock, Service de Génétique Cytogénétique et Embryologie [CHU Pitié-Salpêtrière], CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Department of Clinical Genetics, Leicester Royal Infirmary, University Hospitals Leicester-University Hospitals Leicester, Physiopathologie Cardiovasculaire et Mitochondriale (MITOVASC), Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Kennedy Krieger Institute [Baltimore], Department of Pediatrics, The University of Texas at San Antonio (UTSA), Universiteit Leiden [Leiden], Department of Pathology and Laboratory Medicine [Philadelphia, PA, USA], University of Pennsylvania [Philadelphia]-Perelman School of Medicine, University of Pennsylvania [Philadelphia], Children’s Hospital of Philadelphia (CHOP ), Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Service de génétique clinique, Hôpital Jeanne de Flandre [Lille]-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Ethox Centre, Department of Public Health and Primary Health Care, University of Oxford, Badenoch Building, Old Road Campus, Headington, University of Manchester [Manchester], Central Manchester University Hospitals NHS Foundation Trust, Waseda University, CHU Sainte Justine [Montréal], Cancer Genetics Branch, National Institute of Health (NIH)-National Human Genome Research Institute (NHGRI), and Southampton General Hospital
- Subjects
0301 basic medicine ,Male ,[SDV]Life Sciences [q-bio] ,Developmental Disabilities ,030105 genetics & heredity ,chromatin remodeling ,12p13 ,Neurodevelopmental disorder ,Intellectual disability ,Missense mutation ,Global developmental delay ,Child ,Genetics (clinical) ,Genetics ,Syndrome ,Phenotype ,3. Good health ,intellectual disability ,Child, Preschool ,Female ,medicine.symptom ,Mi-2 Nucleosome Remodeling and Deacetylase Complex ,Adult ,Heart Defects, Congenital ,missense ,Adolescent ,Genotype ,Mutation, Missense ,Biology ,Chromatin remodeling ,Article ,12p13.31 ,03 medical and health sciences ,medicine ,Humans ,ATPase ,Abnormalities, Multiple ,Hearing Loss ,Genetic Association Studies ,Macrocephaly ,Infant, Newborn ,Infant ,medicine.disease ,Chromatin Assembly and Disassembly ,Human genetics ,Megalencephaly ,Musculoskeletal Abnormalities ,030104 developmental biology ,Neurodevelopmental Disorders ,Transcription Factors - Abstract
Sifrim–Hitz–Weiss syndrome (SIHIWES) is a recently described multisystemic neurodevelopmental disorder caused by de novo variants in CHD4. In this study, we investigated the clinical spectrum of the disorder, genotype–phenotype correlations, and the effect of different missense variants on CHD4 function. We collected clinical and molecular data from 32 individuals with mostly de novo variants in CHD4, identified through next-generation sequencing. We performed adenosine triphosphate (ATP) hydrolysis and nucleosome remodeling assays on variants from five different CHD4 domains. The majority of participants had global developmental delay, mild to moderate intellectual disability, brain anomalies, congenital heart defects, and dysmorphic features. Macrocephaly was a frequent but not universal finding. Additional common abnormalities included hypogonadism in males, skeletal and limb anomalies, hearing impairment, and ophthalmic abnormalities. The majority of variants were nontruncating and affected the SNF2-like region of the protein. We did not identify genotype–phenotype correlations based on the type or location of variants. Alterations in ATP hydrolysis and chromatin remodeling activities were observed in variants from different domains. The CHD4-related syndrome is a multisystemic neurodevelopmental disorder. Missense substitutions in different protein domains alter CHD4 function in a variant-specific manner, but result in a similar phenotype in humans.
- Published
- 2020
- Full Text
- View/download PDF
22. Exploring histone loading on HIV DNA reveals a dynamic nucleosome positioning between unintegrated and integrated viral genome
- Author
-
David Depierre, Gaël Petitjean, Heng-Chang Chen, Olivier Cuvier, Monsef Benkirane, Cécile Doyen, Motoki Takaku, Shinichi Machida, and Suzie Thenin-Houssier
- Subjects
Gene Expression Regulation, Viral ,Transcription, Genetic ,Virus Integration ,HIV Infections ,histone ,Genome, Viral ,Biology ,Microbiology ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Transcription (biology) ,Humans ,Nucleosome ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Terminal Repeat Sequences ,virus diseases ,HIV ,Biological Sciences ,Chromatin Assembly and Disassembly ,Long terminal repeat ,Nucleosomes ,3. Good health ,Chromatin ,Cell biology ,Histone ,chemistry ,unintegrated viral DNA ,DNA, Viral ,HIV-1 ,biology.protein ,H3K4me3 ,transcription ,Hypersensitive site ,nucleosome positioning ,030217 neurology & neurosurgery ,DNA - Abstract
Significance The biology of HIV DNA, from its synthesis to its integration into the host genome, remains poorly understood. Here we show that in the nucleus, histones are rapidly loaded on newly synthesized unintegrated HIV DNA. Interestingly, the chromatin architecture around the HIV long terminal repeat (LTR) is different in unintegrated and integrated HIV DNA. Specifically, a nucleosome present only on the DNase hypersensitive site of unintegrated HIV DNA contributes to the transcriptional silencing of unintegrated HIV DNA by preventing RNAPII recruitment., The aim of the present study was to understand the biology of unintegrated HIV-1 DNA and reveal the mechanisms involved in its transcriptional silencing. We found that histones are loaded on HIV-1 DNA after its nuclear import and before its integration in the host genome. Nucleosome positioning analysis along the unintegrated and integrated viral genomes revealed major differences in nucleosome density and position. Indeed, in addition to the well-known nucleosomes Nuc0, Nuc1, and Nuc2 loaded on integrated HIV-1 DNA, we also found NucDHS, a nucleosome that covers the DNase hypersensitive site, in unintegrated viral DNA. In addition, unintegrated viral DNA-associated Nuc0 and Nuc2 were positioned slightly more to the 5′ end relative to their position in integrated DNA. The presence of NucDHS in the proximal region of the long terminal repeat (LTR) promoter was associated with the absence of RNAPII and of the active histone marks H3K4me3 and H3ac at the LTR. Conversely, analysis of integrated HIV-1 DNA showed a loss of NucDHS, loading of RNAPII, and enrichment in active histone marks within the LTR. We propose that unintegrated HIV-1 DNA adopts a repressive chromatin structure that competes with the transcription machinery, leading to its silencing.
- Published
- 2020
- Full Text
- View/download PDF
23. GATA3 mutation disrupts a functional network governed by estrogen receptor, FOXA1 and GATA3
- Author
-
Paul A. Wade, Brian D. Bennett, Bony De Kumar, Motoki Takaku, and Sara A. Grimm
- Subjects
0303 health sciences ,Mutation ,Mutant ,GATA3 ,Estrogen receptor ,Biology ,medicine.disease_cause ,Chromatin ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Nuclear receptor ,030220 oncology & carcinogenesis ,medicine ,FOXA1 ,Transcription factor ,030304 developmental biology - Abstract
Estrogen receptors (ER) are part of the nuclear receptor superfamily of transcription factors and are activated by the steroid hormone 17β-estradiol. ER forms a regulatory network in conjunction with other transcription factors, such as FOXA1 and GATA3. GATA3 has been identified as one of the most frequently mutated genes in breast cancer and is capable of specifying chromatin localization of FOXA1 and ER. How GATA3 mutations impact this transcriptional network is unknown. Here we investigate the function of one of the recurrent patient-derived GATA3 mutations (R330fs) on this regulatory network. Genomic analysis indicates that the R330fs mutant can disrupt the cooperative action of ER, FOXA1, and GATA3, and induce a change in chromatin localization of these factors. Relocalization of ER and FOXA1 is associated with altered chromatin architecture, which leads to differential gene expression in GATA3 mutant cells. These results suggest an active role for GATA3 mutants in ER positive breast tumors.
- Published
- 2019
- Full Text
- View/download PDF
24. Correction: The CHD4-related syndrome: a comprehensive investigation of the clinical spectrum, genotype–phenotype correlations, and molecular basis
- Author
-
Karin Weiss, Hayley P. Lazar, Alina Kurolap, Ariel F. Martinez, Tamar Paperna, Lior Cohen, Marie F. Smeland, Sandra Whalen, Solveig Heide, Boris Keren, Pauline Terhal, Melita Irving, Motoki Takaku, John D. Roberts, Robert M. Petrovich, Samantha A. Schrier Vergano, Amy Kenney, Hanne Hove, Elizabeth DeChene, Shane C. Quinonez, Estelle Colin, Alban Ziegler, Melissa Rumple, Mahim Jain, Danielle Monteil, Elizabeth R. Roeder, Kimberly Nugent, Arie van Haeringen, Michael Gambello, Avni Santani, Līvija Medne, Bryan Krock, Cara M. Skraban, Elaine H. Zackai, Holly A. Dubbs, Thomas Smol, Jamal Ghoumid, Michael J. Parker, Michael Wright, Peter Turnpenny, Jill Clayton-Smith, Kay Metcalfe, Hitoshi Kurumizaka, Bruce D. Gelb, Hagit Baris Feldman, Philippe M. Campeau, Maximilian Muenke, Paul A. Wade, and Katherine Lachlan
- Subjects
Genetics (clinical) - Published
- 2020
- Full Text
- View/download PDF
25. GATA3-dependent cellular reprogramming requires activation-domain dependent recruitment of a chromatin remodeler
- Author
-
Takashi Shimbo, Hitoshi Kurumizaka, Hendrik G. Stunnenberg, Trevor K. Archer, Roberta Menafra, Lalith Perera, Shinichi Machida, Sara A. Grimm, Paul A. Wade, and Motoki Takaku
- Subjects
0301 basic medicine ,Epithelial-Mesenchymal Transition ,Chromosomal Proteins, Non-Histone ,Induced Pluripotent Stem Cells ,Breast Neoplasms ,GATA3 Transcription Factor ,Biology ,Chromatin remodeling ,03 medical and health sciences ,Breast cancer ,GATA3 ,Humans ,Nucleosome ,Pioneer factor ,Molecular Biology ,Transcription factor ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,Genetics ,Research ,Enhancer establishment ,Chromatin binding ,Mesenchymal-to-epithelial transition ,Cellular Reprogramming ,Chromatin Assembly and Disassembly ,Chromatin ,Nucleosomes ,Cell biology ,030104 developmental biology ,Histone ,biology.protein ,Female ,Reprogramming ,Transcription Factors - Abstract
Background Transcription factor-dependent cellular reprogramming is integral to normal development and is central to production of induced pluripotent stem cells. This process typically requires pioneer transcription factors (TFs) to induce de novo formation of enhancers at previously closed chromatin. Mechanistic information on this process is currently sparse. Results Here we explore the mechanistic basis by which GATA3 functions as a pioneer TF in a cellular reprogramming event relevant to breast cancer, the mesenchymal to epithelial transition (MET). In some instances, GATA3 binds previously inaccessible chromatin, characterized by stable, positioned nucleosomes where it induces nucleosome eviction, alters local histone modifications, and remodels local chromatin architecture. At other loci, GATA3 binding induces nucleosome sliding without concomitant generation of accessible chromatin. Deletion of the transactivation domain retains the chromatin binding ability of GATA3 but cripples chromatin reprogramming ability, resulting in failure to induce MET. Conclusions These data provide mechanistic insights into GATA3-mediated chromatin reprogramming during MET, and suggest unexpected complexity to TF pioneering. Successful reprogramming requires stable binding to a nucleosomal site; activation domain-dependent recruitment of co-factors including BRG1, the ATPase subunit of the SWI/SNF chromatin remodeling complex; and appropriate genomic context. The resulting model provides a new conceptual framework for de novo enhancer establishment by a pioneer TF. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-0897-0) contains supplementary material, which is available to authorized users.
- Published
- 2016
- Full Text
- View/download PDF
26. A class of GATA3 mutation reprograms the breast cancer transcriptional network through gain and loss of function
- Author
-
Lalith Perera, Kaliopi Chrysovergis, John Roberts, Brian D. Bennett, Motoki Takaku, Page Myers, Paul A. Wade, Charles M. Perou, Sara A. Grimm, and Charles J. Tucker
- Subjects
Genetics ,0303 health sciences ,Mutation ,GATA3 ,Biology ,medicine.disease_cause ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,030220 oncology & carcinogenesis ,Progesterone receptor ,medicine ,Allele ,Sequence motif ,Gene ,Loss function ,030304 developmental biology - Abstract
GATA3 is frequently mutated in breast cancer; these mutations are widely presumed to be loss of function. Here, we address molecular alterations downstream of a novel class of GATA3 mutations, revealing both gain and loss of function. Mutation of one allele of GATA3 led to loss of binding and decreased expression at a subset of genes, including Progesterone Receptor. At other loci, associated with epithelial to mesenchymal transition, gain of binding at a novel sequence motif correlated with increased gene expression. These results demonstrate that not all GATA3 mutations are equivalent and that these mutations impact breast cancer through gain and loss of function.
- Published
- 2017
- Full Text
- View/download PDF
27. GATA3 zinc finger 2 mutations reprogram the breast cancer transcriptional network
- Author
-
Charles J. Tucker, Paul A. Wade, Sara A. Grimm, Brian D. Bennett, Lalith Perera, Kaliopi Chrysovergis, John Roberts, Page Myers, Charles M. Perou, and Motoki Takaku
- Subjects
0301 basic medicine ,Science ,General Physics and Astronomy ,Mice, Nude ,Breast Neoplasms ,GATA3 Transcription Factor ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,Frameshift mutation ,03 medical and health sciences ,Breast cancer ,Progesterone receptor ,medicine ,Animals ,Humans ,lcsh:Science ,Frameshift Mutation ,Gene ,Zinc finger ,Regulation of gene expression ,Gene Editing ,Mutation ,Mice, Inbred BALB C ,Multidisciplinary ,GATA3 ,Mammary Neoplasms, Experimental ,Zinc Fingers ,General Chemistry ,medicine.disease ,Cellular Reprogramming ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cancer research ,MCF-7 Cells ,lcsh:Q ,Female ,Receptors, Progesterone ,Neoplasm Transplantation - Abstract
GATA3 is frequently mutated in breast cancer; these mutations are widely presumed to be loss-of function despite a dearth of information regarding their effect on disease course or their mechanistic impact on the breast cancer transcriptional network. Here, we address molecular and clinical features associated with GATA3 mutations. A novel classification scheme defines distinct clinical features for patients bearing breast tumors with mutations in the second GATA3 zinc-finger (ZnFn2). An engineered ZnFn2 mutant cell line by CRISPR–Cas9 reveals that mutation of one allele of the GATA3 second zinc finger (ZnFn2) leads to loss of binding and decreased expression at a subset of genes, including Progesterone Receptor. At other loci, associated with epithelial to mesenchymal transition, gain of binding correlates with increased gene expression. These results demonstrate that not all GATA3 mutations are equivalent and that ZnFn2 mutations impact breast cancer through gain and loss-of function., In breast cancer GATA3 is known to be frequently mutated, but the function of these mutations is unclear. Here, the authors utilise CRISPR-Cas9 to model frame-shift mutations in zinc finger 2 of GATA3, highlighting that GATA3 mutation can have gain- or loss-of function effects in breast cancer.
- Published
- 2017
28. Rif1 promotes a repressive chromatin state to safeguard against endogenous retrovirus activation
- Author
-
Guang Hu, Motoki Takaku, Jiajia Wang, Jialun Li, Paul A. Wade, Yufeng Qin, Brian D. Bennett, Li Wang, Jiemin Wong, and Pishun Li
- Subjects
0301 basic medicine ,Telomere-Binding Proteins ,Endogenous retrovirus ,Methylation ,Cell Line ,Histones ,03 medical and health sciences ,Mice ,0302 clinical medicine ,RNA interference ,Genetics ,Animals ,Humans ,Epigenetics ,Cells, Cultured ,Embryonic Stem Cells ,biology ,Endogenous Retroviruses ,Gene regulation, Chromatin and Epigenetics ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,Chromatin ,Cell biology ,030104 developmental biology ,Histone ,HEK293 Cells ,Histone methyltransferase ,DNA methylation ,biology.protein ,Cancer research ,Histone Methyltransferases ,RNA Interference ,Virus Activation ,Reprogramming ,030217 neurology & neurosurgery - Abstract
Transposable elements, including endogenous retroviruses (ERVs), constitute a large fraction of the mammalian genome. They are transcriptionally silenced during early development to protect genome integrity and aberrant transcription. However, the mechanisms that control their repression are not fully understood. To systematically study ERV repression, we carried out an RNAi screen in mouse embryonic stem cells (ESCs) and identified a list of novel regulators. Among them, Rif1 displays the strongest effect. Rif1 depletion by RNAi or gene deletion led to increased transcription and increased chromatin accessibility at ERV regions and their neighboring genes. This transcriptional de-repression becomes more severe when DNA methylation is lost. On the mechanistic level, Rif1 directly occupies ERVs and is required for repressive histone mark H3K9me3 and H3K27me3 assembly and DNA methylation. It interacts with histone methyltransferases and facilitates their recruitment to ERV regions. Importantly, Rif1 represses ERVs in human ESCs as well, and the evolutionally-conserved HEAT-like domain is essential for its function. Finally, Rif1 acts as a barrier during somatic cell reprogramming, and its depletion significantly enhances reprogramming efficiency. Together, our study uncovered many previously uncharacterized repressors of ERVs, and defined an essential role of Rif1 in the epigenetic defense against ERV activation.
- Published
- 2017
29. Exploring histone loading on HIV DNA reveals a dynamic nucleosome positioning between unintegrated and integrated viral genome.
- Author
-
Shinichi Machida, Depierre, David, Heng-Chang Chen, Thenin-Houssier, Suzie, Petitjean, Gaël, Doyen, Cecile M., Motoki Takaku, Cuvier, Olivier, and Benkirane, Monsef
- Subjects
VIRAL genomes ,DNA ,NUCLEAR DNA ,HIV ,HISTONES - Abstract
The aim of the present study was to understand the biology of unintegrated HIV-1 DNA and reveal the mechanisms involved in its transcriptional silencing. We found that histones are loaded on HIV-1 DNA after its nuclear import and before its integration in the host genome. Nucleosome positioning analysis along the unintegrated and integrated viral genomes revealed major differences in nucleosome density and position. Indeed, in addition to the wellknown nucleosomes Nuc0, Nuc1, and Nuc2 loaded on integrated HIV-1 DNA, we also found NucDHS, a nucleosome that covers the DNase hypersensitive site, in unintegrated viral DNA. In addition, unintegrated viral DNA-associated Nuc0 and Nuc2 were positioned slightly more to the 5' end relative to their position in integrated DNA. The presence of NucDHS in the proximal region of the long terminal repeat (LTR) promoter was associated with the absence of RNAPII and of the active histone marks H3K4me3 and H3ac at the LTR. Conversely, analysis of integrated HIV-1 DNA showed a loss of NucDHS, loading of RNAPII, and enrichment in active histone marks within the LTR. We propose that unintegrated HIV-1 DNA adopts a repressive chromatin structure that competes with the transcription machinery, leading to its silencing. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
30. Chromatin architecture may dictate the target site for DMC1, but not for RAD51, during homologous pairing
- Author
-
Hitoshi Kurumizaka, Kazumitsu Maehara, Motoki Takaku, Hiroaki Tachiwana, Shinichi Machida, Yasuyuki Ohkawa, and Wataru Kobayashi
- Subjects
0301 basic medicine ,DNA, Single-Stranded ,Cell Cycle Proteins ,Solenoid (DNA) ,Biology ,Article ,Chromatin remodeling ,Histones ,03 medical and health sciences ,Humans ,Nucleosome ,Histone code ,Homologous Recombination ,ChIA-PET ,Genetics ,Nucleosome binding ,Binding Sites ,Multidisciplinary ,Base Sequence ,Models, Genetic ,fungi ,DNA ,Linker DNA ,Chromatin ,Nucleosomes ,Cell biology ,DNA-Binding Proteins ,Chromosome Pairing ,030104 developmental biology ,Electrophoresis, Polyacrylamide Gel ,Rad51 Recombinase ,Protein Binding - Abstract
In eukaryotes, genomic DNA is compacted as chromatin, in which histones and DNA form the nucleosome as the basic unit. DMC1 and RAD51 are essential eukaryotic recombinases that mediate homologous chromosome pairing during homologous recombination. However, the means by which these two recombinases distinctly function in chromatin have remained elusive. Here we found that, in chromatin, the human DMC1-single-stranded DNA complex bypasses binding to the nucleosome, and preferentially promotes homologous pairing at the nucleosome-depleted regions. Consistently, DMC1 forms ternary complex recombination intermediates with the nucleosome-free DNA or the nucleosome-depleted DNA region. Surprisingly, removal of the histone tails improperly enhances the nucleosome binding by DMC1. In contrast, RAD51 does not specifically target the nucleosome-depleted region in chromatin. These are the first demonstrations that the chromatin architecture specifies the sites to promote the homologous recombination reaction by DMC1, but not by RAD51.
- Published
- 2016
- Full Text
- View/download PDF
31. Biochemical analysis of the human ENA/VASP-family proteins, MENA, VASP and EVL, in homologous recombination
- Author
-
Motoki Takaku, Hiroyuki Ueno, and Hitoshi Kurumizaka
- Subjects
genetic processes ,RAD51 ,macromolecular substances ,Biology ,Biochemistry ,law.invention ,chemistry.chemical_compound ,law ,Homologous chromosome ,Humans ,Homologous Recombination ,Molecular Biology ,Microfilament Proteins ,Ena/Vasp homology proteins ,Actin remodeling ,DNA ,General Medicine ,Surface Plasmon Resonance ,Phosphoproteins ,Molecular biology ,Recombinant Proteins ,Cell biology ,enzymes and coenzymes (carbohydrates) ,chemistry ,Cytoplasm ,health occupations ,Recombinant DNA ,Rad51 Recombinase ,biological phenomena, cell phenomena, and immunity ,Homologous recombination ,Cell Adhesion Molecules - Abstract
MENA, VASP and EVL are members of the ENA/VASP family of proteins and are involved in cytoplasmic actin remodeling. Previously, we found that EVL directly interacts with RAD51, an essential protein in the homologous recombinational repair of double-strand breaks (DSBs) and stimulates the RAD51-mediated recombination reactions in vitro. The EVL-knockdown MCF7 cells exhibited a clear reduction in RAD51-foci formation, suggesting that EVL may function in the DSB repair pathway through RAD51-mediated homologous recombination. However, the DSB repair defects were less significant in the EVL-knockdown cells, implying that two EVL paralogues, MENA and VASP, may complement the EVL function in human cells. Therefore, in the present study, we purified human MENA, VASP and EVL as recombinant proteins, and compared their biochemical activities in vitro. We found that all three proteins commonly exhibited the RAD51 binding, DNA binding and DNA-annealing activities. Stimulation of the RAD51-mediated homologous pairing was also observed with all three proteins. In addition, surface plasmon resonance analyses revealed that MENA, VASP and EVL mutually interacted. These results support the ideas that the ENA/VASP-family proteins are functionally redundant in homologous recombination, and that all three may be involved in the DSB repair pathway in humans.
- Published
- 2011
- Full Text
- View/download PDF
32. Halenaquinone, a chemical compound that specifically inhibits the secondary DNA binding of RAD51
- Author
-
Satoshi Tashiro, Yoichi Nakao, Rob W. M. Van Soest, Takako Ishida-Takaku, Hidekazu Suzuki, Motoki Takaku, Hitoshi Kurumizaka, Shintaro Ishigami, and Takashi Kainuma
- Subjects
chemistry.chemical_classification ,genetic processes ,RAD51 ,Cell Biology ,Biology ,medicine.disease_cause ,Molecular biology ,enzymes and coenzymes (carbohydrates) ,chemistry.chemical_compound ,Enzyme ,chemistry ,Biochemistry ,In vivo ,health occupations ,Genetics ,Nucleic acid ,medicine ,biological phenomena, cell phenomena, and immunity ,Carcinogenesis ,Gene ,DNA ,Function (biology) - Abstract
Mutations and single-nucleotide polymorphisms affecting RAD51 gene function have been identified in several tumors, suggesting that the inappropriate expression of RAD51 activity may cause tumorigenesis. RAD51 is an essential enzyme for the homologous recombinational repair (HRR) of DNA double-strand breaks. In the HRR pathway, RAD51 catalyzes the homologous pairing between single-stranded DNA (ssDNA) and double-stranded DNA (dsDNA), which is the central step of the HRR pathway. To identify a chemical compound that regulates the homologous-pairing activity of RAD51, in the present study, we screened crude extract fractions from marine sponges by the RAD51-mediated homologous-pairing assay. Halenaquinone was identified as an inhibitor of the RAD51 homologous-pairing activity. A surface plasmon resonance analysis indicated that halenaquinone directly bound to RAD51. Intriguingly, halenaquinone specifically inhibited dsDNA binding by RAD51 alone or the RAD51-ssDNA complex, but only weakly affected the RAD51-ssDNA binding. In vivo, halenaquinone significantly inhibited the retention of RAD51 at double-strand break sites. Therefore, halenaquinone is a novel type of RAD51 inhibitor that specifically inhibits the RAD51-dsDNA binding.
- Published
- 2011
- Full Text
- View/download PDF
33. Abstract 5418: GATA3 zinc-finger mutation induces transcriptional reprogramming in breast cancer through gain and loss of function
- Author
-
Paul A. Wade, Sara A. Grimm, Motoki Takaku, John Roberts, Page Myers, Charles M. Perou, and Kaliopi Chrysovergis
- Subjects
Cancer Research ,Mutation ,Mutant ,GATA3 ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Phenotype ,Breast cancer ,Oncology ,Progesterone receptor ,medicine ,Cancer research ,Transcription factor - Abstract
Recent large-scale breast cancer genomic profiling identified frequent mutations in a pioneer transcription factor, GATA3. The expression of GATA3 is a prominent marker of luminal breast tumors, and loss of GATA3 expression is associated with aggressive tumor phenotypes. In addition, GATA3 mutations are frequently found in these luminal tumors. However, the clinical and molecular outcomes of GATA3 mutations are poorly understood. Our novel classification scheme of GATA3 mutations defines distinct clinical features of breast tumors with mutations in the second GATA3 zinc-finger (ZnFn2): frequent observation in luminal B subtype and worse prognosis than other GATA3 mutant cases. To dissect the molecular impacts of these ZnFn2 mutations, we generated a GATA3 mutant breast cancer cell clone by CRISPR-Cas9 gene editing. An engineered ZnFn2 mutant cell line manifested increased tumor growth as xenografts and more aggressive phenotypes in vitro. The ZnFn2 mutation led to loss of GATA3 binding and decreased expression at a subset of genes, including progesterone receptor (PR). The mutant cells also exhibited gain of GATA3 binding at other loci, correlated with increased expression of mesenchymal marker genes. Decreased expression of PR and impaired response to progesterone were crucial for cancer-promoting functions in GATA3 ZnFn2 mutant cells. Downregulation of PR and its downstream genes was also observed in the clinical gene expression data. These results illuminate tumor-promoting functions of GATA3 ZnFn2 mutations in breast cancer. Citation Format: Motoki Takaku, Sara A. Grimm, John D. Roberts, Kaliopi Chrysovergis, Page Myers, Charles M. Perou, Paul A. Wade. GATA3 zinc-finger mutation induces transcriptional reprogramming in breast cancer through gain and loss of function [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5418.
- Published
- 2018
- Full Text
- View/download PDF
34. Single-stranded DNA catenation mediated by human EVL and a type I topoisomerase
- Author
-
Kiyoshi Miyagawa, Motoki Takaku, Shinichi Machida, Hitoshi Kurumizaka, Takehiko Shibata, Noriko Hosoya, Shukuko Ikawa, Hiroyuki Ueno, and Daisuke Takahashi
- Subjects
Topoisomerase ,Actin remodeling ,DNA, Single-Stranded ,Biology ,medicine.disease_cause ,Molecular biology ,DNA, Catenated ,Models, Biological ,chemistry.chemical_compound ,Catenation ,chemistry ,DNA Topoisomerases, Type I ,Genetics ,medicine ,Annealing activity ,biology.protein ,Biophysics ,Humans ,Homologous recombination ,Escherichia coli ,Molecular Biology ,Cell Adhesion Molecules ,Actin ,DNA - Abstract
The human Ena/Vasp-like (EVL) protein is considered to be a bifunctional protein, involved in both actin remodeling and homologous recombination. In the present study, we found that human EVL forms heat-stable multimers of circular single-stranded DNA (ssDNA) molecules in the presence of a type I topoisomerase in vitro. An electron microscopic analysis revealed that the heat-stable ssDNA multimers formed by EVL and topoisomerase were ssDNA catemers. The ssDNA catenation did not occur when either EVL or topoisomerase was omitted from the reaction mixture. A deletion analysis revealed that the ssDNA catenation completely depended on the annealing activity of EVL. Human EVL was captured from a human cell extract by TOPO IIIα-conjugated beads, and the interaction between EVL and TOPO IIIα was confirmed by a surface plasmon resonance analysis. Purified TOPO IIIα catalyzed the ssDNA catenation with EVL as efficiently as the Escherichia coli topoisomerase I. Since the ssDNA cutting and rejoining reactions, which are the sub-steps of ssDNA catenation, may be an essential process in homologous recombination, EVL and TOPO IIIα may function in the processing of DNA intermediates formed during homologous recombination.
- Published
- 2010
35. Holliday junction-binding activity of human SPF45
- Author
-
Naoki Horikoshi, Motoki Takaku, Hitoshi Kurumizaka, Yuichi Morozumi, and Yoshimasa Takizawa
- Subjects
DNA Repair ,DNA repair ,RNA Splicing ,RAD51 ,Cell Cycle Proteins ,Biology ,chemistry.chemical_compound ,Genetics ,Holliday junction ,Humans ,Replication protein A ,Recombination, Genetic ,DNA, Cruciform ,RNA recognition motif ,Mutagenesis ,DNA ,Cell Biology ,Molecular biology ,Cell biology ,DNA-Binding Proteins ,chemistry ,Gene Targeting ,Rad51 Recombinase ,Homologous recombination - Abstract
SPF45 is considered to be a bifunctional protein that functions in splicing and DNA repair. A previous genetic study reported that Drosophila SPF45 participates in the DNA-repair pathway with a RAD51-family protein, RAD201, suggesting that SPF45 may function in DNA repair by the homologous-recombination pathway. To study the function of SPF45 in homologous recombination, we purified human SPF45 and found that it preferentially binds to the Holliday junction, which is a key DNA intermediate in the homologous-recombination pathway. Deletion analyses revealed that the RNA recognition motif, which is located in the C-terminal region of human SPF45, is not involved in DNA binding. On the other hand, alanine-scanning mutagenesis identified the N-terminal lysine residues, which may be involved in Holliday junction binding by human SPF45. We also found that human SPF45 significantly binds to a RAD51 paralog, RAD51B, although it also binds to RAD51 and DMC1 with lower affinity. These biochemical results support the idea that human SPF45 functions in DNA repair by homologous recombination.
- Published
- 2010
- Full Text
- View/download PDF
36. Biochemical analysis of the human EVL domains in homologous recombination
- Author
-
Daisuke Takahashi, Shugo Nakayama, Hitoshi Kurumizaka, Motoki Takaku, and Shinichi Machida
- Subjects
RAD51 ,Cell Biology ,Plasma protein binding ,Biology ,Biochemistry ,Molecular biology ,chemistry.chemical_compound ,chemistry ,EVH1 domain ,Homologous chromosome ,Homologous recombination ,Molecular Biology ,Recombination ,Function (biology) ,DNA - Abstract
EVL is a member of the ENA/VASP family, which is involved in actin-remodeling processes. Previously, we reported that human EVL directly interacts with RAD51, which is an essential protein in the homologous recombinational repair of DNA double-strand breaks, and stimulates RAD51-mediated recombination reactions in vitro. To identify the EVL domain required for the recombination function, we purified the EVL fragments EVL(1-221) and EVL(222-418), which contain the EVH1 and Pro-rich domains and the EVH2 domain, respectively. We found that EVL(222-418) possesses DNA-binding and RAD51-binding activities, and also stimulates RAD51-mediated homologous pairing. In contrast, EVL(1-221) did not exhibit any of these activities. Therefore, the EVH2 domain, which is highly conserved among the ENA/VASP family proteins, may be responsible for the recombination function of EVL. Structured digital abstract: * MINT-7239394: EVL (uniprotkb:Q9UI08) binds (MI:0407) to RAD51 (uniprotkb:Q06609) by pull down (MI:0096).
- Published
- 2009
- Full Text
- View/download PDF
37. The ATR-Chk1 pathway plays a role in the generation of centrosome aberrations induced by Rad51C dysfunction
- Author
-
Motoki Takaku, Yoshitaka Tomoda, Aiko Kinomura, Takanori Tsuruga, Hitoshi Kurumizaka, Miyuki Okajima, Kiyoshi Miyagawa, Mari Katsura, Hiromu K. Mishima, Takashi Yoshihara, Osamu Date, and Mari Ishida
- Subjects
animal structures ,Mutant ,RAD51 ,Aneuploidy ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Genome Integrity, Repair and Replication ,Protein Serine-Threonine Kinases ,Biology ,RNA interference ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Gene silencing ,Centrosome ,Recombination, Genetic ,medicine.disease ,DNA-Binding Proteins ,Checkpoint Kinase 1 ,Cancer research ,RAD51C ,RNA Interference ,biological phenomena, cell phenomena, and immunity ,Homologous recombination ,Protein Kinases - Abstract
Rad51C is a central component of two complexes formed by five Rad51 paralogs in vertebrates. These complexes are involved in repairing DNA double-strand breaks through homologous recombination. Despite accumulating evidence suggesting that the paralogs may prevent aneuploidy by controlling centrosome integrity, Rad51C's role in maintaining chromosome stability remains unclear. Here we demonstrate that Rad51C deficiency leads to both centrosome aberrations in an ATR-Chk1-dependent manner and increased aneuploidy in human cells. While it was reported that Rad51C deficiency did not cause centrosome aberrations in interphase in hamster cells, such aberrations were observed in interphase in HCT116 cells with Rad51C dysfunction. Caffeine treatment and down-regulation of ATR, but not that of ATM, reduced the frequency of centrosome aberrations in the mutant cells. Silencing of Rad51C by RNA interference in HT1080 cells resulted in similar aberrations. Treatment with a Chk1 inhibitor and silencing of Chk1 also reduced the frequency in HCT116 mutants. Accumulation of Chk1 at the centrosome and nuclear foci of gamma H2AX were increased in the mutants. Moreover, the mutant cells had a higher frequency of aneuploidy. These findings indicate that the ATR-Chk1 pathway plays a role in increased centrosome aberrations induced by Rad51C dysfunction.
- Published
- 2009
- Full Text
- View/download PDF
38. Relaxed Chromatin Formation and Weak Suppression of Homologous Pairing by the Testis-Specific Linker Histone H1T
- Author
-
Ryota Hayashida, Satoshi Tashiro, Jiying Sun, Aiko Kinomura, Motoki Takaku, Atsuhiko Fukuto, Hitoshi Kurumizaka, and Shinichi Machida
- Subjects
0301 basic medicine ,Histone-modifying enzymes ,RAD51 ,Biochemistry ,Cell Line ,Histones ,03 medical and health sciences ,Nucleosome ,Histone code ,Humans ,Recombination, Genetic ,030102 biochemistry & molecular biology ,biology ,DNA Helicases ,Nuclear Proteins ,Molecular biology ,Linker DNA ,Chromatin ,Cell biology ,Nucleosomes ,DNA-Binding Proteins ,030104 developmental biology ,Histone ,Histone methyltransferase ,biology.protein ,Rad51 Recombinase - Abstract
Linker histones bind to nucleosomes and compact polynucleosomes into a higher-order chromatin configuration. Somatic and germ cell-specific linker histone subtypes have been identified and may have distinct functions. In this study, we reconstituted polynucleosomes containing human histones H1.2 and H1T, as representative somatic and germ cell-specific linker histones, respectively, and found that H1T forms less compacted chromatin, as compared to H1.2. An in vitro homologous pairing assay revealed that H1T weakly inhibited RAD51/RAD54-mediated homologous pairing in chromatin, although the somatic H1 subtypes, H1.0, H1.1, H1.2, H1.3, H1.4, and H1.5, substantially suppressed it. An in vivo recombination assay revealed that H1T overproduction minimally affected the recombination frequency, but significant suppression was observed when H1.2 was overproduced in human cells. These results suggested that the testis-specific linker histone, H1T, possesses a specific function to produce the chromatin architecture required for proper chromosome regulation, such as homologous recombination.
- Published
- 2016
39. GATA3 in Breast Cancer: Tumor Suppressor or Oncogene?
- Author
-
Motoki Takaku, Paul A. Wade, and Sara A. Grimm
- Subjects
Mammary gland ,Breast Neoplasms ,GATA3 Transcription Factor ,Biology ,medicine.disease_cause ,Article ,law.invention ,Breast cancer ,law ,Genetics ,medicine ,Humans ,Genes, Tumor Suppressor ,Molecular Biology ,Transcription factor ,Mutation ,Oncogene ,GATA3 ,Cancer ,Oncogenes ,medicine.disease ,medicine.anatomical_structure ,Immunology ,Cancer research ,Suppressor ,Female - Abstract
GATA3 is a highly conserved, essential transcription factor expressed in a number of tissues, including the mammary gland. GATA3 expression is required for normal development of the mammary gland where it is estimated to be the most abundant transcription factor in luminal epithelial cells. In breast cancer, GATA3 expression is highly correlated with the luminal transcriptional program. Recent genomic analysis of human breast cancers has revealed high-frequency mutation in GATA3 in luminal tumors, suggesting “driver” function(s). Here we discuss mutation of GATA3 in breast cancer and the potential mechanism(s) by which mutation may lead to a growth advantage in cancer.
- Published
- 2015
40. Nap1 stimulates homologous recombination by RAD51 and RAD54 in higher-ordered chromatin containing histone H1
- Author
-
Satoshi Tashiro, Kiyoe Ura, Atsuhiko Fukuto, Masae Ikura, Hitoshi Kurumizaka, Ryo Matsuda, Aiko Kinomura, Akihisa Osakabe, Yasunori Horikoshi, Hiroaki Tachiwana, Tsuyoshi Ikura, Wataru Kobayashi, Shinichi Machida, Motoki Takaku, Hidekazu Suzuki, and Jiying Sun
- Subjects
DNA Repair ,RAD51 ,Article ,Chromatin remodeling ,Cell Line ,Histones ,Histone H1 ,Histone H2A ,Histone methylation ,Escherichia coli ,Humans ,Nucleosome ,Histone code ,Homologous Recombination ,Adenosine Triphosphatases ,tRNA Methyltransferases ,Multidisciplinary ,biology ,fungi ,DNA Helicases ,Nuclear Proteins ,Proteins ,Molecular biology ,Chromatin ,Nucleosomes ,Cell biology ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,DNA Repair Enzymes ,Histone ,biology.protein ,Rad51 Recombinase - Abstract
Homologous recombination plays essential roles in mitotic DNA double strand break (DSB) repair and meiotic genetic recombination. In eukaryotes, RAD51 promotes the central homologous-pairing step during homologous recombination, but is not sufficient to overcome the reaction barrier imposed by nucleosomes. RAD54, a member of the ATP-dependent nucleosome remodeling factor family, is required to promote the RAD51-mediated homologous pairing in nucleosomal DNA. In higher eukaryotes, most nucleosomes form higher-ordered chromatin containing the linker histone H1. However, the mechanism by which RAD51/RAD54-mediated homologous pairing occurs in higher-ordered chromatin has not been elucidated. In this study, we found that a histone chaperone, Nap1, accumulates on DSB sites in human cells and DSB repair is substantially decreased in Nap1-knockdown cells. We determined that Nap1 binds to RAD54, enhances the RAD54-mediated nucleosome remodeling by evicting histone H1 and eventually stimulates the RAD51-mediated homologous pairing in higher-ordered chromatin containing histone H1.
- Published
- 2014
- Full Text
- View/download PDF
41. Human PSF concentrates DNA and stimulates duplex capture in DMC1-mediated homologous pairing
- Author
-
Shinichiro Chuma, Motoki Takaku, Ryohei Ino, Mihoko Hosokawa, Hitoshi Kurumizaka, and Yuichi Morozumi
- Subjects
Male ,Mitotic crossover ,DNA repair ,DNA, Single-Stranded ,Cell Cycle Proteins ,Biology ,chemistry.chemical_compound ,Mice ,Transcription (biology) ,Genetics ,Recombinase ,Animals ,Humans ,Homologous Recombination ,PTB-Associated Splicing Factor ,Molecular Biology ,Activator (genetics) ,fungi ,Nuclear Proteins ,RNA-Binding Proteins ,DNA ,Phosphate-Binding Proteins ,Molecular biology ,Spermatozoa ,Cell biology ,DNA-Binding Proteins ,chemistry ,DMC1 ,Rad51 Recombinase ,Homologous recombination - Abstract
PSF is considered to have multiple functions in RNA processing, transcription and DNA repair by mitotic recombination. In the present study, we found that PSF is produced in spermatogonia, spermatocytes and spermatids, suggesting that PSF may also function in meiotic recombination. We tested the effect of PSF on homologous pairing by the meiosis-specific recombinase DMC1, and found that human PSF robustly stimulated it. PSF synergistically enhanced the formation of a synaptic complex containing DMC1, ssDNA and dsDNA during homologous pairing. The PSF-mediated DMC1 stimulation may be promoted by its DNA aggregation activity, which increases the local concentrations of ssDNA and dsDNA for homologous pairing by DMC1. These results suggested that PSF may function as an activator for the meiosis-specific recombinase DMC1 in higher eukaryotes.
- Published
- 2011
42. Halenaquinone, a chemical compound that specifically inhibits the secondary DNA binding of RAD51
- Author
-
Motoki, Takaku, Takashi, Kainuma, Takako, Ishida-Takaku, Shintaro, Ishigami, Hidekazu, Suzuki, Satoshi, Tashiro, Rob W M, van Soest, Yoichi, Nakao, and Hitoshi, Kurumizaka
- Subjects
DNA-Binding Proteins ,Sequence Homology, Nucleic Acid ,Quinones ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Rad51 Recombinase ,Complex Mixtures ,In Vitro Techniques ,Phosphoinositide-3 Kinase Inhibitors ,Porifera - Abstract
Mutations and single-nucleotide polymorphisms affecting RAD51 gene function have been identified in several tumors, suggesting that the inappropriate expression of RAD51 activity may cause tumorigenesis. RAD51 is an essential enzyme for the homologous recombinational repair (HRR) of DNA double-strand breaks. In the HRR pathway, RAD51 catalyzes the homologous pairing between single-stranded DNA (ssDNA) and double-stranded DNA (dsDNA), which is the central step of the HRR pathway. To identify a chemical compound that regulates the homologous-pairing activity of RAD51, in the present study, we screened crude extract fractions from marine sponges by the RAD51-mediated homologous-pairing assay. Halenaquinone was identified as an inhibitor of the RAD51 homologous-pairing activity. A surface plasmon resonance analysis indicated that halenaquinone directly bound to RAD51. Intriguingly, halenaquinone specifically inhibited dsDNA binding by RAD51 alone or the RAD51-ssDNA complex, but only weakly affected the RAD51-ssDNA binding. In vivo, halenaquinone significantly inhibited the retention of RAD51 at double-strand break sites. Therefore, halenaquinone is a novel type of RAD51 inhibitor that specifically inhibits the RAD51-dsDNA binding.
- Published
- 2011
43. GEMIN2 promotes accumulation of RAD51 at double-strand breaks in homologous recombination
- Author
-
Kouji Hirota, Masayuki Takahashi, Takehiko Shibata, Hitoshi Kurumizaka, Toshiaki Kogame, Motoki Takaku, Yoshimasa Takizawa, Yong Qing, Shunichi Takeda, Takashi Tsujita, Yuichi Morozumi, and Takako Ishida
- Subjects
Spliceosome ,DNA Repair ,DNA damage ,DNA repair ,genetic processes ,RAD51 ,Biology ,Genome Integrity, Repair and Replication ,Cell Line ,chemistry.chemical_compound ,Genetics ,Homologous chromosome ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Replication protein A ,Cell Proliferation ,Recombination, Genetic ,SMN Complex Proteins ,DNA ,Molecular biology ,Cell biology ,enzymes and coenzymes (carbohydrates) ,chemistry ,health occupations ,Rad51 Recombinase ,biological phenomena, cell phenomena, and immunity ,Homologous recombination ,Chickens - Abstract
RAD51 is a key factor in homologous recombination (HR) and plays an essential role in cellular proliferation by repairing DNA damage during replication. The assembly of RAD51 at DNA damage is strictly controlled by RAD51 mediators, including BRCA1 and BRCA2. We found that human RAD51 directly binds GEMIN2/SIP1, a protein involved in spliceosome biogenesis. Biochemical analyses indicated that GEMIN2 enhances the RAD51-DNA complex formation by inhibiting RAD51 dissociation from DNA, and thereby stimulates RAD51-mediated homologous pairing. GEMIN2 also enhanced the RAD51-mediated strand exchange, when RPA was pre-bound to ssDNA before the addition of RAD51. To analyze the function of GEMIN2, we depleted GEMIN2 in the chicken DT40 line and in human cells. The loss of GEMIN2 reduced HR efficiency and resulted in a significant decrease in the number of RAD51 subnuclear foci, as observed in cells deficient in BRCA1 and BRCA2. These observations and our biochemical analyses reveal that GEMIN2 regulates HR as a novel RAD51 mediator.
- Published
- 2010
44. Biochemical analysis of the human EVL domains in homologous recombination
- Author
-
Motoki, Takaku, Shinichi, Machida, Shugo, Nakayama, Daisuke, Takahashi, and Hitoshi, Kurumizaka
- Subjects
Recombination, Genetic ,Humans ,Rad51 Recombinase ,Cell Adhesion Molecules ,Protein Binding ,Protein Structure, Tertiary - Abstract
EVL is a member of the ENA/VASP family, which is involved in actin-remodeling processes. Previously, we reported that human EVL directly interacts with RAD51, which is an essential protein in the homologous recombinational repair of DNA double-strand breaks, and stimulates RAD51-mediated recombination reactions in vitro. To identify the EVL domain required for the recombination function, we purified the EVL fragments EVL(1-221) and EVL(222-418), which contain the EVH1 and Pro-rich domains and the EVH2 domain, respectively. We found that EVL(222-418) possesses DNA-binding and RAD51-binding activities, and also stimulates RAD51-mediated homologous pairing. In contrast, EVL(1-221) did not exhibit any of these activities. Therefore, the EVH2 domain, which is highly conserved among the ENA/VASP family proteins, may be responsible for the recombination function of EVL. Structured digital abstract: * MINT-7239394: EVL (uniprotkb:Q9UI08) binds (MI:0407) to RAD51 (uniprotkb:Q06609) by pull down (MI:0096).
- Published
- 2009
45. Recombination Activator Function of the Novel RAD51- and RAD51B-binding Protein, Human EVL*S⃞
- Author
-
Noriko Hosoya, Shugo Nakayama, Yoshimasa Takizawa, Shinichi Machida, Hitoshi Kurumizaka, Kiyoshi Miyagawa, Isao Sakane, Motoki Takaku, and Takehiko Shibata
- Subjects
RAD51 ,Plasma protein binding ,Biology ,Biochemistry ,DNA-binding protein ,chemistry.chemical_compound ,Cell Line, Tumor ,Homologous chromosome ,Humans ,Molecular Biology ,Recombination, Genetic ,Activator (genetics) ,Binding protein ,Cell Biology ,DNA ,DNA repair protein XRCC4 ,Molecular biology ,DNA-Binding Proteins ,Chromosome Pairing ,chemistry ,Gene Knockdown Techniques ,DNA: Replication, Repair, Recombination, and Chromosome Dynamics ,Rad51 Recombinase ,Cell Adhesion Molecules ,Protein Binding - Abstract
The RAD51 protein is a central player in homologous recombinational repair. The RAD51B protein is one of five RAD51 paralogs that function in the homologous recombinational repair pathway in higher eukaryotes. In the present study, we found that the human EVL (Ena/Vasp-like) protein, which is suggested to be involved in actin-remodeling processes, unexpectedly binds to the RAD51 and RAD51B proteins and stimulates the RAD51-mediated homologous pairing and strand exchange. The EVL knockdown cells impaired RAD51 assembly onto damaged DNA after ionizing radiation or mitomycin C treatment. The EVL protein alone promotes single-stranded DNA annealing, and the recombination activities of the EVL protein are further enhanced by the RAD51B protein. The expression of the EVL protein is not ubiquitous, but it is significantly expressed in breast cancer-derived MCF7 cells. These results suggest that the EVL protein is a novel recombination factor that may be required for repairing specific DNA lesions, and that may cause tumor malignancy by its inappropriate expression.
- Published
- 2009
46. Human PSF binds to RAD51 and modulates its homologous-pairing and strand-exchange activities
- Author
-
Hitoshi Kurumizaka, Yoshimasa Takizawa, Yuichi Morozumi, and Motoki Takaku
- Subjects
Base pair ,genetic processes ,RAD51 ,DNA, Single-Stranded ,RNA-binding protein ,Biology ,Binding, Competitive ,chemistry.chemical_compound ,Transcription (biology) ,Genetics ,Recombinase ,Humans ,PTB-Associated Splicing Factor ,Base Pairing ,Molecular Biology ,Recombination, Genetic ,Binding Sites ,RNA ,RNA-Binding Proteins ,DNA ,Molecular biology ,Cell biology ,Protein Structure, Tertiary ,enzymes and coenzymes (carbohydrates) ,chemistry ,health occupations ,Rad51 Recombinase ,biological phenomena, cell phenomena, and immunity ,Homologous recombination - Abstract
RAD51, a eukaryotic recombinase, catalyzes homologous-pairing and strand-exchange reactions, which are essential steps in homologous recombination and recombinational repair of double strand breaks. On the other hand, human PSF was originally identified as a component of spliceosomes, and its multiple functions in RNA processing, transcription and DNA recombination were subsequently revealed. In the present study, we found that PSF directly interacted with RAD51. PSF significantly enhanced RAD51-mediated homologous pairing and strand exchange at low RAD51 concentrations; however, in contrast, it inhibited these RAD51-mediated recombination reactions at the optimal RAD51 concentration. Deletion analyses revealed that the N-terminal region of PSF possessed the RAD51- and DNA-binding activities, but the central region containing the RNA-recognition motifs bound neither RAD51 nor DNA. These results suggest that PSF may have dual functions in homologous recombination and RNA processing through its N-terminal and central regions, respectively.
- Published
- 2009
47. Abstract 964: GATA3 modulates chromatin structure to establish active enhancers in breast cancer cells
- Author
-
Paul A. Wade, Hitoshi Kurumizaka, Lalith Perera, Motoki Takaku, Sara A. Grimm, Shinichi Machida, and Takashi Shimbo
- Subjects
Cancer Research ,Oncology ,Chromatin binding ,Pioneer factor ,Cancer research ,FOXA1 ,Biology ,Transcription factor ,ChIA-PET ,Chromatin remodeling ,Chromatin ,Epithelial cell differentiation - Abstract
Master transcription factors regulate cell-type-specific gene expression to define cellular identities. One such gene, GATA3, is a key regulator of multiple cellular programs, including T lymphocyte development, mammary luminal epithelial cell differentiation and trophoblast development. Recently, comprehensive genomic analysis has identified GATA3 as one of the most frequently mutated genes in breast cancer. It is also known that GATA3 expression levels directly correlate with favorable prognosis. These findings strongly suggest that GATA3 plays a critical role in tumorigenesis. However, the molecular mechanism(s) underlying GATA3-mediated gene regulation in breast cancer cells is not clearly defined. GATA3 participates in a complicated regulatory network with FOXA1 and ER-alpha, governing the transcriptional program in luminal tumors. Biochemical analyses indicate that: (1) GATA3 binds to chromatin in an estrogen-independent manner, (2) GATA3 acts upstream of FOXA1. These studies suggest GATA3 may act as a pioneer factor, which is capable of independently associating with closed chromatin and modulating chromatin structure to establish an active enhancer. In order to investigate GATA3 function as a pioneer transcription factor, we chose the MDA-MB-231 breast cancer cell line, which is GATA3, FOXA1 and ER-alpha negative, and established stable cell lines expressing wild-type GATA3 or GFP as a control. Consistent with previous results, GATA3-expressing cells represented an epithelial phenotype at the cellular and molecular level. To determine whether GATA3 can direct reprogramming of chromatin conformation, we performed genome-wide analyses of the chromatin binding activity of GATA3 and its impact on histone modifications and chromatin structure. We will present recent results describing how GATA3 licenses enhancer function to direct the luminal transcriptional program. Citation Format: Motoki Takaku, Sara A. Grimm, Takashi Shimbo, Lalith Perera, Shinichi Machida, Hitoshi Kurumizaka, Paul A. Wade. GATA3 modulates chromatin structure to establish active enhancers in breast cancer cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 964. doi:10.1158/1538-7445.AM2015-964
- Published
- 2015
- Full Text
- View/download PDF
48. Rif1 promotes a repressive chromatin state to safeguard against endogenous retrovirus activation.
- Author
-
Pishun Li, Li Wang, Bennett, Brian D., Wang, Jiajia, Jialun Li, Yufeng Qin, Motoki Takaku, Wade, Paul A., Jiemin Wong, and Guang Hu
- Published
- 2017
- Full Text
- View/download PDF
49. Abstract 475: GATA3 mutations in breast cancer
- Author
-
Sara A. Grimm, Shimbo Takashi, Aleksandra B. Adomas, Motoki Takaku, and Paul A. Wade
- Subjects
Genetics ,Cancer Research ,GATA3 ,Cancer ,Biology ,medicine.disease ,Metastasis ,Frameshift mutation ,Exon ,Breast cancer ,Oncology ,medicine ,Cancer research ,Ectopic expression ,Estrogen receptor alpha - Abstract
Breast cancer is one of the most common cancers in women worldwide, resulting in over 400,000 deaths per year. Approximately 90% of breast cancers occur in women who have no family history of breast cancer, indicating that most breast cancers occur following spontaneous mutations resulting from the aging process or the action of environmental agents. Breast cancers can be divided into several subtypes base on gene expression profiles. Luminal breast cancer, the most common subtype, is characterized by expression of estrogen receptor alpha (ER-alpha). The zinc finger transcription factor GATA3, which is known as a regulator of mammary gland development, is frequently expressed in luminal breast cancer cells, and its expression is highly correlated with ER-alpha. In breast cancers, the expression levels of GATA3 directly correlate with favorable prognosis, and its ectopic expression leads to the suppression of tumor metastasis. Comprehensive genomic analysis of breast tumors has revealed frequent somatic mutations of GATA3 in luminal breast cancers. However it is unclear how these GATA3 mutations affect the biological properties of breast cancer. These mutations are located at exclusively in exons 5 and 6 that encode the carboxyl terminus of the protein. Exon 5 contains the second of two zinc finger domains (ZnF2), which is important for the DNA binding activity of GATA3. Exon 6 has no previously described biochemical function. In this study, we focus on two mutations found in cancer patients - a frame shift at tyrosine 345 (Y345fs) and a frame shift at arginine 330 (R330fs). These two mutants are of particular interest as they produce truncated proteins, and both mutants lack exon 6. Y345fs preserves ZnF2, while R330fs interrupts ZnF2. These two mutants permit dissection of the molecular outcomes flowing from loss of exon 6 (Y345fs) versus the combination of loss of ZnF2 and exon 6 (R330fs). To assess the impact of these mutations, we established breast cancer cells expressing Y345fs and R330fs respectively, and analyzed their phenotype at the cellular and molecular levels. These studies begin to address the biochemical and functional outcomes of commonly occurring mutations in breast cancer. Citation Format: Motoki Takaku, Aleksandra Adomas, Sara A. Grimm, Shimbo Takashi, Paul A. Wade. GATA3 mutations in breast cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 475. doi:10.1158/1538-7445.AM2014-475
- Published
- 2014
- Full Text
- View/download PDF
50. Breast tumor specific mutation in GATA3 affects physiological mechanisms regulating transcription factor turnover
- Author
-
Jennifer K. Sims, Sara A. Grimm, Christine C. Malone, Aleksandra B. Adomas, Paul A. Wade, and Motoki Takaku
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Cancer Research ,Transcription, Genetic ,Estrogen receptor ,Breast Neoplasms ,GATA3 Transcription Factor ,Biology ,Frameshift mutation ,Breast cancer ,GATA3 ,Genetics ,Humans ,skin and connective tissue diseases ,Transcription factor ,Protein Stability ,Estrogen Receptor alpha ,Estrogens ,Chromatin ,Cell biology ,ChIP-seq ,Oncology ,Mutation ,Cancer research ,MCF-7 Cells ,Female ,FOXA1 ,Estrogen receptor alpha ,Chromatin immunoprecipitation ,Research Article - Abstract
Background The transcription factor GATA3 is a favorable prognostic indicator in estrogen receptor-α (ERα)-positive breast tumors in which it participates with ERα and FOXA1 in a complex transcriptional regulatory program driving tumor growth. GATA3 mutations are frequent in breast cancer and have been classified as driver mutations. To elucidate the contribution(s) of GATA3 alterations to cancer, we studied two breast cancer cell lines, MCF7, which carries a heterozygous frameshift mutation in the second zinc finger of GATA3, and T47D, wild-type at this locus. Methods Immunofluorescence staining and subcellular fractionation were employed to verify cellular localization of GATA3 in T47D and MCF7 cells. To test protein stability, cells were treated with translation inhibitor, cycloheximide or proteasome inhibitor, MG132, and GATA3 abundance was measured over time using immunoblot. GATA3 turn-over in response to hormone was determined by treating the cells with estradiol or ERα agonist, ICI 182,780. DNA binding ability of recombinant GATA3 was evaluated using electrophoretic mobility shift assay and heparin chromatography. Genomic location of GATA3 in MCF7 and T47D cells was assessed by chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-seq). Results GATA3 localized in the nucleus in T47D and MCF7 cells, regardless of the mutation status. The truncated protein in MCF7 had impaired interaction with chromatin and was easily released from the nucleus. Recombinant mutant GATA3 was able to bind DNA to a lesser degree than the wild-type protein. Heterozygosity for the truncating mutation conferred protection from regulated turnover of GATA3, ERα and FOXA1 following estrogen stimulation in MCF7 cells. Thus, mutant GATA3 uncoupled protein-level regulation of master regulatory transcription factors from hormone action. Consistent with increased protein stability, ChIP-seq profiling identified greater genome-wide accumulation of GATA3 in MCF7 cells bearing the mutation, albeit with a similar distribution across the genome, comparing to T47D cells. Conclusions We propose that this specific, cancer-derived mutation in GATA3 deregulates physiologic protein turnover, stabilizes GATA3 binding across the genome and modulates the response of breast cancer cells to estrogen signaling.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.