1
|
Liang X, Liu X, Zhang L, Liu J, Yan R, Li H, Zeng X, Wang H. Targeting the Ajuba/Notch axis increases the sensitivity of colon cancer cells to 5-fluorouracil. Cytojournal 2024; 21:44. [PMID: 39737130 PMCID: PMC11683402 DOI: 10.25259/cytojournal_44_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/09/2024] [Indexed: 01/01/2025] Open
Abstract
Objective Colorectal cancer is severely challenging because of the insufficient understanding of the mechanism underlying its resistance to clinical chemotherapy. The purpose of our study is to investigate the role of the LIM protein Ajuba (JUB) in the chemoresistance of colon cancer and its potential effect on clinical treatment. Material and Methods The protein levels of JUB in colon cancer tissues were evaluated using Western blot analysis and immunohistochemistry assays. The correlation between JUB and the prognosis of patients with colorectal cancer was determined using Kaplan-Meier plot analysis. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays were employed to determine the 50% inhibitory concentration of 5-fluorouracil (5-FU) and thus assess the effect of JUB on the effectiveness of 5-FU. In addition, the rate of cellular apoptosis was measured using fluorescence-activated cell sorting assays. Side population and sphere formation analyses were conducted to determine the role of JUB in promoting the stem cell-like traits of colon cancer cells. In vivo assays were performed and detect whether the downregulation of JUB induces 5-FU sensitivity. Moreover, luciferase and Western blot assays were employed to uncover the mechanism through which JUB promotes chemoresistance in colon cancer. Results JUB expression was upregulated in chemoresistant colon cancer (P < 0.001) and correlated with relapse-free survival (P = 0.000002). Functionally, the upregulation of JUB conferred 5-FU resistance to colon cancer cells in vitro, whereas the downregulation of JUB induced 5-FU sensitivity in colon cancer cells in vivo. The high expression of JUB promoted the tumorigenic capability of colon cancer cells. Furthermore, the increased expression of JUB activated multiple downstream genes of the Notch signaling pathway with increased expression in JUB-overexpressing cells but reduced expression in JUB-silenced cells. Importantly, the inhibition of Notch signaling using a small-molecule inhibitor significantly suppressed JUB-induced chemoresistance. Conclusion Results suggest that JUB plays an important role and may serve as a biomarker for the clinical treatment of patients with 5-FU-resistant colon cancer.
Collapse
Affiliation(s)
- Xinghua Liang
- The First Clinical Medical College, Jinan University, Guangzhou, China
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People`s Hospital of Guangzhou), Guangzhou, China
| | - Xuelian Liu
- Department of Gastroenterology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Long Zhang
- Department of Endoscopy Center, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Junhao Liu
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Rong Yan
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People`s Hospital of Guangzhou), Guangzhou, China
| | - Haiyan Li
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People`s Hospital of Guangzhou), Guangzhou, China
| | - Xiancheng Zeng
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Hong Wang
- The First Clinical Medical College, Jinan University, Guangzhou, China
- Department of Gastroenterology, Guangzhou First People`s Hospital, Guangzhou, China
| |
Collapse
|
2
|
Goswami P, Banks CA, Thornton J, Bengs B, Sardiu ME, Florens L, Washburn MP. Distinct regions within SAP25 recruit O-linked glycosylation, DNA demethylation, and ubiquitin ligase and hydrolase activities to the Sin3/HDAC complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583553. [PMID: 38496433 PMCID: PMC10942353 DOI: 10.1101/2024.03.05.583553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Epigenetic control of gene expression is crucial for maintaining gene regulation. Sin3 is an evolutionarily conserved repressor protein complex mainly associated with histone deacetylase (HDAC) activity. A large number of proteins are part of Sin3/HDAC complexes, and the function of most of these members remains poorly understood. SAP25, a previously identified Sin3A associated protein of 25 kDa, has been proposed to participate in regulating gene expression programs involved in the immune response but the exact mechanism of this regulation is unclear. SAP25 is not expressed in HEK293 cells, which hence serve as a natural knockout system to decipher the molecular functions uniquely carried out by this Sin3/HDAC subunit. Using molecular, proteomic, protein engineering, and interaction network approaches, we show that SAP25 interacts with distinct enzymatic and regulatory protein complexes in addition to Sin3/HDAC. While the O-GlcNAc transferase (OGT) and the TET1 /TET2/TET3 methylcytosine dioxygenases have been previously linked to Sin3/HDAC, in HEK293 cells, these interactions were only observed in the affinity purification in which an exogenously expressed SAP25 was the bait. Additional proteins uniquely recovered from the Halo-SAP25 pull-downs included the SCF E3 ubiquitin ligase complex SKP1/FBXO3/CUL1 and the ubiquitin carboxyl-terminal hydrolase 11 (USP11), which have not been previously associated with Sin3/HDAC. Finally, we use mutational analysis to demonstrate that distinct regions of SAP25 participate in its interaction with USP11, OGT/TETs, and SCF(FBXO3).) These results suggest that SAP25 may function as an adaptor protein to coordinate the assembly of different enzymatic complexes to control Sin3/HDAC-mediated gene expression.
Collapse
Affiliation(s)
- Pratik Goswami
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Charles A.S. Banks
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
| | - Janet Thornton
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Bethany Bengs
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mihaela E. Sardiu
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Laurence Florens
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
| | - Michael P. Washburn
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| |
Collapse
|
3
|
García de Herreros A. Dual role of Snail1 as transcriptional repressor and activator. Biochim Biophys Acta Rev Cancer 2024; 1879:189037. [PMID: 38043804 DOI: 10.1016/j.bbcan.2023.189037] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
Snail1 transcriptional factor plays a key role in the control of epithelial to mesenchymal transition, a process that remodels tumor cells increasing their invasion and chemo-resistance as well as reprograms their metabolism and provides stemness properties. During this transition, Snail1 acts as a transcriptional repressor and, as growing evidences have demonstrated, also as a direct activator of mesenchymal genes. In this review, I describe the different proteins that interact with Snail1 and are responsible for these two different functions on gene expression; I focus on the transcriptional factors that associate to Snail1 in their target promoters, both activated and repressed. I also present working models for Snail1 action both as repressor and activator and raise some issues that still need to be investigated.
Collapse
Affiliation(s)
- Antonio García de Herreros
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Unidad Asociada al CSIC, Barcelona, Spain; Departament de Medicina i Ciències de la Vida, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
4
|
Frank D, Patnana PK, Vorwerk J, Mao L, Gopal LM, Jung N, Hennig T, Ruhnke L, Frenz JM, Kuppusamy M, Autry R, Wei L, Sun K, Mohammed Ahmed HM, Künstner A, Busch H, Müller H, Hutter S, Hoermann G, Liu L, Xie X, Al-Matary Y, Nimmagadda SC, Cano FC, Heuser M, Thol F, Göhring G, Steinemann D, Thomale J, Leitner T, Fischer A, Rad R, Röllig C, Altmann H, Kunadt D, Berdel WE, Hüve J, Neumann F, Klingauf J, Calderon V, Opalka B, Dührsen U, Rosenbauer F, Dugas M, Varghese J, Reinhardt HC, von Bubnoff N, Möröy T, Lenz G, Batcha AMN, Giorgi M, Selvam M, Wang E, McWeeney SK, Tyner JW, Stölzel F, Mann M, Jayavelu AK, Khandanpour C. Germ line variant GFI1-36N affects DNA repair and sensitizes AML cells to DNA damage and repair therapy. Blood 2023; 142:2175-2191. [PMID: 37756525 PMCID: PMC10733838 DOI: 10.1182/blood.2022015752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/06/2023] [Accepted: 07/24/2023] [Indexed: 09/29/2023] Open
Abstract
ABSTRACT Growth factor independence 1 (GFI1) is a DNA-binding transcription factor and a key regulator of hematopoiesis. GFI1-36N is a germ line variant, causing a change of serine (S) to asparagine (N) at position 36. We previously reported that the GFI1-36N allele has a prevalence of 10% to 15% among patients with acute myeloid leukemia (AML) and 5% to 7% among healthy Caucasians and promotes the development of this disease. Using a multiomics approach, we show here that GFI1-36N expression is associated with increased frequencies of chromosomal aberrations, mutational burden, and mutational signatures in both murine and human AML and impedes homologous recombination (HR)-directed DNA repair in leukemic cells. GFI1-36N exhibits impaired binding to N-Myc downstream-regulated gene 1 (Ndrg1) regulatory elements, causing decreased NDRG1 levels, which leads to a reduction of O6-methylguanine-DNA-methyltransferase (MGMT) expression levels, as illustrated by both transcriptome and proteome analyses. Targeting MGMT via temozolomide, a DNA alkylating drug, and HR via olaparib, a poly-ADP ribose polymerase 1 inhibitor, caused synthetic lethality in human and murine AML samples expressing GFI1-36N, whereas the effects were insignificant in nonmalignant GFI1-36S or GFI1-36N cells. In addition, mice that received transplantation with GFI1-36N leukemic cells treated with a combination of temozolomide and olaparib had significantly longer AML-free survival than mice that received transplantation with GFI1-36S leukemic cells. This suggests that reduced MGMT expression leaves GFI1-36N leukemic cells particularly vulnerable to DNA damage initiating chemotherapeutics. Our data provide critical insights into novel options to treat patients with AML carrying the GFI1-36N variant.
Collapse
Affiliation(s)
- Daria Frank
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Pradeep Kumar Patnana
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University Cancer Center Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Jan Vorwerk
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | - Lianghao Mao
- Proteomics and Cancer Cell Signaling Group, Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center and Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Lavanya Mokada Gopal
- Proteomics and Cancer Cell Signaling Group, Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center and Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Noelle Jung
- Proteomics and Cancer Cell Signaling Group, Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center and Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Thorben Hennig
- Proteomics and Cancer Cell Signaling Group, Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center and Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Leo Ruhnke
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Joris Maximillian Frenz
- Proteomics and Cancer Cell Signaling Group, Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center and Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Maithreyan Kuppusamy
- Proteomics and Cancer Cell Signaling Group, Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center and Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Robert Autry
- Hopp Children’s Cancer Center, Heidelberg, Germany
| | - Lanying Wei
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Kaiyan Sun
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | - Helal Mohammed Mohammed Ahmed
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University Cancer Center Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | | | | | | | - Longlong Liu
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Department of Hematology, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoqing Xie
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yahya Al-Matary
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Subbaiah Chary Nimmagadda
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University Cancer Center Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Fiorella Charles Cano
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Felicitas Thol
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Gudrun Göhring
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Doris Steinemann
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Jürgen Thomale
- Institute of Cell Biology, University Hospital Essen, Essen, Germany
| | - Theo Leitner
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University Cancer Center Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Anja Fischer
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technische Universität München, Munich, Germany
- Center for Translational Cancer Research, School of Medicine, Technische Universität München, Munich, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technische Universität München, Munich, Germany
- Center for Translational Cancer Research, School of Medicine, Technische Universität München, Munich, Germany
- Department of Medicine II, Klinikum Rechts der Isar, School of Medicine, Technische Universität München, Munich, Germany
| | | | | | | | - Wolfgang E. Berdel
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | - Jana Hüve
- Fluorescence Microscopy Facility Münster, Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Felix Neumann
- Fluorescence Microscopy Facility Münster, Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
- Refined Laser Systems GmbH, Münster, Germany
| | - Jürgen Klingauf
- Fluorescence Microscopy Facility Münster, Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Virginie Calderon
- Bioinformatic Core Facility, Institut de Recherches Cliniques de Montréal, Montréal, QC, Canada
| | - Bertram Opalka
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Frank Rosenbauer
- Institute of Molecular Tumor Biology, Faculty of Medicine, University of Münster, Münster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University Cancer Center Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Tarik Möröy
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | - Aarif M. N. Batcha
- Institute of Medical Data Processing, Biometrics and Epidemiology, Faculty of Medicine, Ludwig Maximilians University Munich, Munich, Germany
- Data Integration for Future Medicine, Ludwig Maximilian University Munich, Munich, Germany
| | - Marianna Giorgi
- Roswell Park Comprehensive Cancer Center, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY
| | - Murugan Selvam
- Roswell Park Comprehensive Cancer Center, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY
| | - Eunice Wang
- Roswell Park Comprehensive Cancer Center, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY
| | - Shannon K. McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR
| | - Jeffrey W. Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR
| | - Friedrich Stölzel
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
- Department of Medicine II, Division for Stem Cell Transplantation and Cellular Immunotherapy, University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein Kiel, Christian Albrecht University Kiel, Kiel, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Munich, Germany
| | - Ashok Kumar Jayavelu
- Proteomics and Cancer Cell Signaling Group, Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center and Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
- Hopp Children’s Cancer Center, Heidelberg, Germany
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Munich, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory and Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Cyrus Khandanpour
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, University Cancer Center Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| |
Collapse
|
5
|
Wang H, Lin Z, Nian Z, Zhang W, Liu W, Yan F, Xiao Z, Wang X, Zhang Z, Ma Z, Liu Z. Hematopoietic transcription factor GFI1 promotes anchorage independence by sustaining ERK activity in cancer cells. J Clin Invest 2022; 132:149551. [PMID: 35819844 PMCID: PMC9433100 DOI: 10.1172/jci149551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/07/2022] [Indexed: 11/26/2022] Open
Abstract
The switch from anchorage-dependent to anchorage-independent growth is essential for epithelial metastasis. The underlying mechanism, however, is not fully understood. In this study, we identified growth factor independent-1 (GFI1), a transcription factor that drives the transition from adherent endothelial cells to suspended hematopoietic cells during hematopoiesis, as a critical regulator of anchorage independence in lung cancer cells. GFI1 elevated the numbers of circulating and lung-infiltrating tumor cells in xenograft models and predicted poor prognosis of patients with lung cancer. Mechanistically, GFI1 inhibited the expression of multiple adhesion molecules and facilitated substrate detachment. Concomitantly, GFI1 reconfigured the chromatin structure of the RASGRP2 gene and increased its expression, causing Rap1 activation and subsequent sustained ERK activation upon detachment, and this led to ERK signaling dependency in tumor cells. Our studies unveiled a mechanism by which carcinoma cells hijacked a hematopoietic factor to gain anchorage independence and suggested that the intervention of ERK signaling may suppress metastasis and improve the therapeutic outcome of patients with GFI1-positive lung cancer.
Collapse
Affiliation(s)
- Hao Wang
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Zhenzhen Lin
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Zhe Nian
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Wei Zhang
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Wenxu Liu
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Fei Yan
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Zengtuan Xiao
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xia Wang
- Department of Gastrointestinal Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhenfa Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhenyi Ma
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Zhe Liu
- Department of Immunology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
6
|
Fraszczak J, Arman KM, Lacroix M, Vadnais C, Gaboury L, Möröy T. Severe Inflammatory Reactions in Mice Expressing a GFI1 P2A Mutant Defective in Binding to the Histone Demethylase KDM1A (LSD1). THE JOURNAL OF IMMUNOLOGY 2021; 207:1599-1615. [PMID: 34408010 DOI: 10.4049/jimmunol.2001146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 07/07/2021] [Indexed: 11/19/2022]
Abstract
GFI1 is a DNA-binding transcription factor that regulates hematopoiesis by repressing target genes through its association with complexes containing histone demethylases such as KDM1A (LSD1) and histone deacetylases (HDACs). To study the consequences of the disruption of the complex between GFI1 and histone-modifying enzymes, we have used knock-in mice harboring a P2A mutation in GFI1 coding region that renders it unable to bind LSD1 and associated histone-modifying enzymes such as HDACs. GFI1P2A mice die prematurely and show increased numbers of memory effector and regulatory T cells in the spleen accompanied by a severe systemic inflammation with high serum levels of IL-6, TNF-α, and IL-1β and overexpression of the gene encoding the cytokine oncostatin M (OSM). We identified lung alveolar macrophages, CD8 T cell from the spleen and thymic eosinophils, and monocytes as the sources of these cytokines in GFI1P2A mice. Chromatin immunoprecipitation showed that GFI1/LSD1 complexes occupy sites at the Osm promoter and an intragenic region of the Tnfα gene and that a GFI1P2A mutant still remains bound at these sites even without LSD1. Methylation and acetylation of histone H3 at these sites were enriched in cells from GFI1P2A mice, the H3K27 acetylation being the most significant. These data suggest that the histone modification facilitated by GFI1 is critical to control inflammatory pathways in different cell types, including monocytes and eosinophils, and that a disruption of GFI1-associated complexes can lead to systemic inflammation with fatal consequences.
Collapse
Affiliation(s)
| | - Kaifee Mohammad Arman
- Institut de Recherches Cliniques de Montréal, Montreal, Canada.,Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Marion Lacroix
- Institut de Recherches Cliniques de Montréal, Montreal, Canada.,Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Charles Vadnais
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
| | - Louis Gaboury
- Unité de Recherche en Histologie et Pathologie Moléculaire, Institut de Recherche en Immunologie et en Cancérologie, Montreal, Canada.,Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, Montreal, Canada; and
| | - Tarik Möröy
- Institut de Recherches Cliniques de Montréal, Montreal, Canada; .,Division of Experimental Medicine, McGill University, Montreal, Canada.,Département de Microbiologie Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montreal, Canada
| |
Collapse
|
7
|
The transcription factors GFI1 and GFI1B as modulators of the innate and acquired immune response. Adv Immunol 2021; 149:35-94. [PMID: 33993920 DOI: 10.1016/bs.ai.2021.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
GFI1 and GFI1B are small nuclear proteins of 45 and 37kDa, respectively, that have a simple two-domain structure: The first consists of a group of six c-terminal C2H2 zinc finger motifs that are almost identical in sequence and bind to very similar, specific DNA sites. The second is an N-terminal 20 amino acid SNAG domain that can bind to the pocket of the histone demethylase KDM1A (LSD1) near its active site. When bound to DNA, both proteins act as bridging factors that bring LSD1 and associated proteins into the vicinity of methylated substrates, in particular histone H3 or TP53. GFI1 can also bring methyl transferases such as PRMT1 together with its substrates that include the DNA repair proteins MRE11 and 53BP1, thereby enabling their methylation and activation. While GFI1B is expressed almost exclusively in the erythroid and megakaryocytic lineage, GFI1 has clear biological roles in the development and differentiation of lymphoid and myeloid immune cells. GFI1 is required for lymphoid/myeloid and monocyte/granulocyte lineage decision as well as the correct nuclear interpretation of a number of important immune-signaling pathways that are initiated by NOTCH1, interleukins such as IL2, IL4, IL5 or IL7, by the pre TCR or -BCR receptors during early lymphoid differentiation or by T and B cell receptors during activation of lymphoid cells. Myeloid cells also depend on GFI1 at both stages of early differentiation as well as later stages in the process of activation of macrophages through Toll-like receptors in response to pathogen-associated molecular patterns. The knowledge gathered on these factors over the last decades puts GFI1 and GFI1B at the center of many biological processes that are critical for both the innate and acquired immune system.
Collapse
|
8
|
Beauchemin H, Möröy T. Multifaceted Actions of GFI1 and GFI1B in Hematopoietic Stem Cell Self-Renewal and Lineage Commitment. Front Genet 2020; 11:591099. [PMID: 33193732 PMCID: PMC7649360 DOI: 10.3389/fgene.2020.591099] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/23/2020] [Indexed: 12/15/2022] Open
Abstract
Growth factor independence 1 (GFI1) and the closely related protein GFI1B are small nuclear proteins that act as DNA binding transcriptional repressors. Both recognize the same consensus DNA binding motif via their C-terminal zinc finger domains and regulate the expression of their target genes by recruiting chromatin modifiers such as histone deacetylases (HDACs) and demethylases (LSD1) by using an N-terminal SNAG domain that comprises only 20 amino acids. The only region that is different between both proteins is the region that separates the zinc finger domains and the SNAG domain. Both proteins are co-expressed in hematopoietic stem cells (HSCs) and, to some extent, in multipotent progenitors (MPPs), but expression is specified as soon as early progenitors and show signs of lineage bias. While expression of GFI1 is maintained in lymphoid primed multipotent progenitors (LMPPs) that have the potential to differentiate into both myeloid and lymphoid cells, GFI1B expression is no longer detectable in these cells. By contrast, GFI1 expression is lost in megakaryocyte precursors (MKPs) and in megakaryocyte-erythrocyte progenitors (MEPs), which maintain a high level of GFI1B expression. Consequently, GFI1 drives myeloid and lymphoid differentiation and GFI1B drives the development of megakaryocytes, platelets, and erythrocytes. How such complementary cell type- and lineage-specific functions of GFI1 and GFI1B are maintained is still an unresolved question in particular since they share an almost identical structure and very similar biochemical modes of actions. The cell type-specific accessibility of GFI1/1B binding sites may explain the fact that very similar transcription factors can be responsible for very different transcriptional programming. An additional explanation comes from recent data showing that both proteins may have additional non-transcriptional functions. GFI1 interacts with a number of proteins involved in DNA repair and lack of GFI1 renders HSCs highly susceptible to DNA damage-induced death and restricts their proliferation. In contrast, GFI1B binds to proteins of the beta-catenin/Wnt signaling pathway and lack of GFI1B leads to an expansion of HSCs and MKPs, illustrating the different impact that GFI1 or GFI1B has on HSCs. In addition, GFI1 and GFI1B are required for endothelial cells to become the first blood cells during early murine development and are among those transcription factors needed to convert adult endothelial cells or fibroblasts into HSCs. This role of GFI1 and GFI1B bears high significance for the ongoing effort to generate hematopoietic stem and progenitor cells de novo for the autologous treatment of blood disorders such as leukemia and lymphoma.
Collapse
Affiliation(s)
| | - Tarik Möröy
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
9
|
Dommann N, Sánchez-Taltavull D, Eggs L, Birrer F, Brodie T, Salm L, Baier FA, Medová M, Humbert M, Tschan MP, Beldi G, Candinas D, Stroka D. The LIM Protein Ajuba Augments Tumor Metastasis in Colon Cancer. Cancers (Basel) 2020; 12:cancers12071913. [PMID: 32679899 PMCID: PMC7409172 DOI: 10.3390/cancers12071913] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer, along with its high potential for recurrence and metastasis, is a major health burden. Uncovering proteins and pathways required for tumor cell growth is necessary for the development of novel targeted therapies. Ajuba is a member of the LIM domain family of proteins whose expression is positively associated with numerous cancers. Our data shows that Ajuba is highly expressed in human colon cancer tissue and cell lines. Publicly available data from The Cancer Genome Atlas shows a negative correlation between survival and Ajuba expression in patients with colon cancer. To investigate its function, we transduced SW480 human colon cancer cells, with lentiviral constructs to knockdown or overexpress Ajuba protein. The transcriptome of the modified cell lines was analyzed by RNA sequencing. Among the pathways enriched in the differentially expressed genes, were cell proliferation, migration and differentiation. We confirmed our sequencing data with biological assays; cells depleted of Ajuba were less proliferative, more sensitive to irradiation, migrated less and were less efficient in colony formation. In addition, loss of Ajuba expression decreased the tumor burden in a murine model of colorectal metastasis to the liver. Taken together, our data supports that Ajuba promotes colon cancer growth, migration and metastasis and therefore is a potential candidate for targeted therapy.
Collapse
Affiliation(s)
- Noëlle Dommann
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Daniel Sánchez-Taltavull
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Linda Eggs
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Fabienne Birrer
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Tess Brodie
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Lilian Salm
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Felix Alexander Baier
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Michaela Medová
- Department of Biomedical Research, Radiation Oncology, University of Bern, Bern University Hospital, 3008 Bern, Switzerland;
| | - Magali Humbert
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (M.H.); (M.P.T.)
| | - Mario P. Tschan
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (M.H.); (M.P.T.)
| | - Guido Beldi
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Daniel Candinas
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Deborah Stroka
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
- Correspondence: ; Tel.: +41-31-632-27-48
| |
Collapse
|
10
|
Li R, Li P, Xing W, Qiu H. Heterogeneous genomic aberrations in esophageal squamous cell carcinoma: a review. Am J Transl Res 2020; 12:1553-1568. [PMID: 32509161 PMCID: PMC7269976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 04/25/2020] [Indexed: 06/11/2023]
Abstract
Esophageal cancer (EC) causes hundreds of thousands of deaths a year worldwide, especially the major subtype esophageal squamous cell carcinoma (ESCC). With the advent of next-generation sequencing and the availability of commercial microarrays, abnormities in genetic levels have been revealed in various independent researches. High frequencies of structure variations (SVs), single nucleotide variations (SNVs) and copy-number alterations (CNAs) in ESCCs are uncovered, and ESCC shows high levels of inter- and intratumor heterogeneity, implying diverse evolutionary trajectories. This review tries to explain the pathogenesis of ESCC on the scope of most often mutated genes based on prior studies, hopes to offer some hints for diagnosis and therapy in clinic.
Collapse
Affiliation(s)
- Renling Li
- Quality and Standards Academy, Shenzhen Technology UniversityShenzhen 518060, China
| | - Peng Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhou 450008, China
| | - Wenqun Xing
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhou 450008, China
| | - Huiling Qiu
- Quality and Standards Academy, Shenzhen Technology UniversityShenzhen 518060, China
| |
Collapse
|
11
|
Ajuba: An emerging signal transducer in oncogenesis. Pharmacol Res 2019; 151:104546. [PMID: 31740385 DOI: 10.1016/j.phrs.2019.104546] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/05/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022]
Abstract
The LIM protein Ajuba contains an unstructured proline/glycine-rich preLIM region in the N terminus and conserved tandem LIM motifs in the C terminus. Additionally, Ajuba contains both nuclear export sequences (NES) and nuclear localization sequences (NLS), which enable Ajuba shuttle between the cytoplasm and the nucleus. Thus, Ajuba can act as a versatile scaffold participating in assembly of variety of protein complexes to execute multiple cellular functions including cell adhesion, motility, mitosis, survival, gene expression, microRNA processing and mechanical force sensing. Numerous studies have demonstrated that Ajuba plays important roles in oncogenesis and progression by regulating major signalling pathways such as Wnt, RAS/ERK, JAK/STAT and Hippo, and by acting as a co-regulator of key transcription factors such as Snail, Sp1 and nuclear hormone receptors. Clinically, Ajuba is highly expressed in various types of tumors and can be a marker for prognosis and diagnosis. In this review, we aim to summarize the up-to-date literatures on the signaling pathways mediated by Ajuba and its associated protein complexes in oncogenesis, and to discuss Ajuba as a potential target for new therapeutics to treat cancers.
Collapse
|
12
|
Möröy T, Khandanpour C. Role of GFI1 in Epigenetic Regulation of MDS and AML Pathogenesis: Mechanisms and Therapeutic Implications. Front Oncol 2019; 9:824. [PMID: 31508375 PMCID: PMC6718700 DOI: 10.3389/fonc.2019.00824] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/12/2019] [Indexed: 01/12/2023] Open
Abstract
Growth factor independence 1 (GFI1) is a DNA binding zinc finger protein, which can mediate transcriptional repression mainly by recruiting histone-modifying enzymes to its target genes. GFI1 plays important roles in hematopoiesis, in particular by regulating both the function of hematopoietic stem- and precursor cells and differentiation along myeloid and lymphoid lineages. In recent years, a number of publications have provided evidence that GFI1 is involved in the pathogenesis of acute myeloid leukemia (AML), its proposed precursor, myelodysplastic syndrome (MDS), and possibly also in the progression from MDS to AML. For instance, expression levels of the GFI1 gene correlate with patient survival and treatment response in both AML and MDS and can influence disease progression and maintenance in experimental animal models. Also, a non-synonymous single nucleotide polymorphism (SNP) of GFI1, GFI1-36N, which encodes a variant GFI1 protein with a decreased efficiency to act as a transcriptional repressor, was found to be a prognostic factor for the development of AML and MDS. Both the GFI1-36N variant as well as reduced expression of the GFI1 gene lead to genome-wide epigenetic changes at sites where GFI1 occupies target gene promoters and enhancers. These epigenetic changes alter the response of leukemic cells to epigenetic drugs such as HDAC- or HAT inhibitors, indicating that GFI1 expression levels and genetic variants of GFI1 are of clinical relevance. Based on these and other findings, specific therapeutic approaches have been proposed to treat AML by targeting some of the epigenetic changes that occur as a consequence of GFI1 expression. Here, we will review the well-known role of Gfi1 as a transcription factor and describe the more recently discovered functions of GFI1 that are independent of DNA binding and how these might affect disease progression and the choice of epigenetic drugs for therapeutic regimens of AML and MDS.
Collapse
Affiliation(s)
- Tarik Möröy
- Department of Hematopoiesis and Cancer, Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Cyrus Khandanpour
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| |
Collapse
|
13
|
Zhang B, Song L, Cai J, Li L, Xu H, Li M, Wang J, Shi M, Chen H, Jia H, Hou Z. The LIM protein Ajuba/SP1 complex forms a feed forward loop to induce SP1 target genes and promote pancreatic cancer cell proliferation. J Exp Clin Cancer Res 2019; 38:205. [PMID: 31101117 PMCID: PMC6525466 DOI: 10.1186/s13046-019-1203-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The aim of this study is to explore the molecular mechanism of the LIM protein Ajuba and the transcription factor SP1 in the pathogenesis and progression of PDAC. Ajuba is a newly defined transcriptional co-regulator and plays important role in various cancer development, while SP1 is a classic transcription factor, and is closely related with a variety of gene expression and cancer development including PDAC. METHODS The expression of Ajuba and SP1 in PDAC tissues was detected by immunohistochemistry (IHC), and the correlation between expression level and clinical prognosis of Ajuba and SP1 was extensively analyzed using online tools. The interaction between Ajuba and SP1 was examined by co-immunoprecipitation (co-IP) and GST-pulldown assays. Stable cell lines were established via lentiviral infection, and was examined by qRT-PCR and western blot assays. The effects of Ajuba/SP1 on PDAC cell proliferation were examined using CCK8 and colony formation assays. Luciferase reporter and chromatin immunoprecipitation (ChIP) assays were employed to examine the transcription activity. RESULTS The expression level (protein and mRNA) of Ajuba and SP1 was elevated in PDAC tissues and was positively correlated; patients with high Ajuba and SP1 expression had a poor prognosis. Mechanistically, Ajuba binds to the C-terminus of SP1 and functions as a co-activator to enhance SP1 gene expression and promote cell proliferation; the promoter of Ajuba contains functional SP1 responsive elements and Ajuba itself is a target gene of SP1. CONCLUSION Ajuba functions as a co-activator of SP1 to induce its target gene, and that Ajuba itself is a target genes of SP1. Ajuba/SP1 complex could form a feed forward loop to drive SP1 target gene transcription and promote cell proliferation of pancreatic cancer cells. Ajuba and SP1 might be biomarkers for PDAC diagnostics, prognosis and targets for new therapeutics.
Collapse
Affiliation(s)
- Bosen Zhang
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025 China
- Hongqiao International Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Liwei Song
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiali Cai
- Department of Radiology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003 China
| | - Lei Li
- Department of Thoracic Surgery, Lanling People’s Hospital, Lanling County, Linyi, 277700 China
| | - Hong Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Mengying Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Jiamin Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Minmin Shi
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025 China
| | - Hao Chen
- Department of Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025 China
| | - Hao Jia
- Hongqiao International Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Zhaoyuan Hou
- Hongqiao International Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
- Department of Thoracic Surgery, Lanling People’s Hospital, Lanling County, Linyi, 277700 China
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| |
Collapse
|
14
|
Tang Q, Xu M, Xu J, Xie X, Yang H, Gan L. Gfi1-GCE inducible Cre line for hair cell-specific gene manipulation in mouse inner ear. Genesis 2019; 57:e23304. [PMID: 31077553 DOI: 10.1002/dvg.23304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 01/16/2023]
Abstract
Tissue-specific inducible Cre recombinase mouse lines allow precise genetic manipulations in spatiotemporal manners and are pivotal for functional studies of genes during development and in adults. Growth factor independence 1 (GFI1) is an essential transcription factor expressed in the hair cells of mouse inner ear and Gfi1 locus serves as an excellent anchor site to drive the expression of inducible Cre recombinase in mouse inner hair cells. In this study, we have generated Gfi1-P2A-GFP-CreERT2 (Gfi1-GCE) knock-in mouse line by in-frame fusion of a self-cleaving GCE to the C-terminus of GFI1. We have shown that as predicted, the expression of GCE and GFI1 was detected specifically in the cytosol and nuclei of hair cells, respectively, of uninduced Gfi1-GCE mice, suggesting the successful cleavage and simultaneous expression of GFI1 and GCE. In addition, the in-frame fusion of the self-cleaving GCE does not interrupt the function of Gfi1 in the inner ear. Administration of tamoxifen leads to nuclear translocation of GCE and results in an efficient activation of tdTomato reporter gene expression specifically in most hair cells throughout development and in adults. Thus, this inducible Gfi1-GCE mouse line is a highly efficient Cre deleter and is suitable for gene manipulation in developing and adult inner ear hair cells.
Collapse
Affiliation(s)
- Qi Tang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York
| | - Mei Xu
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York.,Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jiadong Xu
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York.,Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaoling Xie
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York
| | - Hua Yang
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Gan
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, New York
| |
Collapse
|
15
|
Lsd1 as a therapeutic target in Gfi1-activated medulloblastoma. Nat Commun 2019; 10:332. [PMID: 30659187 PMCID: PMC6338772 DOI: 10.1038/s41467-018-08269-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/21/2018] [Indexed: 01/22/2023] Open
Abstract
Drugs that modify the epigenome are powerful tools for treating cancer, but these drugs often have pleiotropic effects, and identifying patients who will benefit from them remains a major clinical challenge. Here we show that medulloblastomas driven by the transcription factor Gfi1 are exquisitely dependent on the enzyme lysine demethylase 1 (Kdm1a/Lsd1). We demonstrate that Lsd1 physically associates with Gfi1, and that these proteins cooperate to inhibit genes involved in neuronal commitment and differentiation. We also show that Lsd1 is essential for Gfi1-mediated transformation: Gfi1 proteins that cannot recruit Lsd1 are unable to drive tumorigenesis, and genetic ablation of Lsd1 markedly impairs tumor growth in vivo. Finally, pharmacological inhibitors of Lsd1 potently inhibit growth of Gfi1-driven tumors. These studies provide important insight into the mechanisms by which Gfi1 contributes to tumorigenesis, and identify Lsd1 inhibitors as promising therapeutic agents for Gfi1-driven medulloblastoma. Medulloblastoma is one of the most prevalent malignant brain tumors in children and has very poor prognosis. In this study, the authors show, using a mouse model of medulloblastoma, that Gfi1 promotes tumor growth by recruiting Lsd1, that this interaction inhibits genes involved in neuronal differentiation, and that Lsd1 may be a therapeutic target in Gfi1-activated tumors.
Collapse
|
16
|
Reduced expression but not deficiency of GFI1 causes a fatal myeloproliferative disease in mice. Leukemia 2018; 33:110-121. [PMID: 29925903 PMCID: PMC6326955 DOI: 10.1038/s41375-018-0166-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/25/2018] [Accepted: 05/10/2018] [Indexed: 12/15/2022]
Abstract
Growth factor independent 1 (Gfi1) controls myeloid differentiation by regulating gene expression and limits the activation of p53 by facilitating its de-methylation at Lysine 372. In human myeloid leukemia, low GFI1 levels correlate with an inferior prognosis. Here, we show that knockdown (KD) of Gfi1 in mice causes a fatal myeloproliferative disease (MPN) that could progress to leukemia after additional mutations. Both KO and KD mice accumulate myeloid cells that show signs of metabolic stress and high levels of reactive oxygen species. However, only KO cells have elevated levels of Lysine 372 methylated p53. This suggests that in contrast to absence of GFI1, KD of GFI1 leads to the accumulation of myeloid cells because sufficient amount of GFI1 is present to impede p53-mediated cell death, leading to a fatal MPN. The combination of myeloid accumulation and the ability to counteract p53 activity under metabolic stress could explain the role of reduced GF1 expression in human myeloid leukemia.
Collapse
|
17
|
Cheng B, Tang S, Zhe N, Ma D, Yu K, Wei D, Zhou Z, Lu T, Wang J, Fang Q. Low expression of GFI-1 Gene is associated with Panobinostat-resistance in acute myeloid leukemia through influencing the level of HO-1. Biomed Pharmacother 2018; 100:509-520. [DOI: 10.1016/j.biopha.2018.02.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/11/2018] [Accepted: 02/13/2018] [Indexed: 02/07/2023] Open
|
18
|
Adamik J, Silbermann R, Marino S, Sun Q, Anderson JL, Zhou D, Xie XQ, Roodman GD, Galson DL. XRK3F2 Inhibition of p62-ZZ Domain Signaling Rescues Myeloma-Induced GFI1-Driven Epigenetic Repression of the Runx2 Gene in Pre-osteoblasts to Overcome Differentiation Suppression. Front Endocrinol (Lausanne) 2018; 9:344. [PMID: 30008697 PMCID: PMC6033965 DOI: 10.3389/fendo.2018.00344] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/07/2018] [Indexed: 01/05/2023] Open
Abstract
Multiple myeloma bone disease (MMBD) is characterized by non-healing lytic bone lesions that persist even after a patient has achieved a hematologic remission. We previously reported that p62 (sequestosome-1) in bone marrow stromal cells (BMSC) is critical for the formation of MM-induced signaling complexes that mediate OB suppression. Importantly, XRK3F2, an inhibitor of the p62-ZZ domain, blunted MM-induced Runx2 suppression in vitro, and induced new bone formation and remodeling in the presence of tumor in vivo. Additionally, we reported that MM cells induce the formation of repressive chromatin on the Runx2 gene in BMSC via direct binding of the transcriptional repressor GFI1, which recruits the histone modifiers, histone deacetylase 1 (HDAC1) and Enhancer of zeste homolog 2 (EZH2). In this study we investigated the mechanism by which blocking p62-ZZ domain-dependent signaling prevents MM-induced suppression of Runx2 in BMSC. XRK3F2 prevented MM-induced upregulation of Gfi1 and repression of the Runx2 gene when present in MM-preOB co-cultures. We also show that p62-ZZ-domain blocking by XRK3F2 also prevented MM conditioned media and TNF plus IL7-mediated Gfi1 mRNA upregulation and the concomitant Runx2 repression, indicating that XRK3F2's prevention of p62-ZZ domain signaling within preOB is involved in the response. Chromatin immunoprecipitation (ChIP) analyses revealed that XRK3F2 decreased MM-induced GFI1 occupancy at the Runx2-P1 promoter and prevented recruitment of HDAC1, thus preserving the transcriptionally permissive chromatin mark H3K9ac on Runx2 and allowing osteogenic differentiation. Furthermore, treatment of MM-exposed preOB with XRK3F2 after MM removal decreased GFI1 enrichment at Runx2-P1 and rescued MM-induced suppression of Runx2 mRNA and its downstream osteogenic gene targets together with increased osteogenic differentiation. Further, primary BMSC (hBMSC) from MM patients (MM-hBMSC) had little ability to increase H3K9ac on the Runx2 promoter in osteogenic conditions when compared to hBMSC from healthy donors (HD). XRK3F2 treatment enriched Runx2 gene H3K9ac levels in MM-hBMSC to the level observed in HD-hBMSC, but did not alter HD-hBMSC H3K9ac. Importantly, XRK3F2 treatment of long-term MM-hBMSC cultures rescued osteogenic differentiation and mineralization. Our data show that blocking p62-ZZ domain-dependent signaling with XRK3F2 can reverse epigenetic-based mechanisms of MM-induced Runx2 suppression and promote osteogenic differentiation.
Collapse
Affiliation(s)
- Juraj Adamik
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rebecca Silbermann
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Silvia Marino
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Quanhong Sun
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Judith L. Anderson
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Dan Zhou
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - G. David Roodman
- Division of Hematology-Oncology, Department of Medicine, Indiana University, Indianapolis, IN, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, United States
| | - Deborah L. Galson
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- *Correspondence: Deborah L. Galson ;
| |
Collapse
|
19
|
Bi L, Ma F, Tian R, Zhou Y, Lan W, Song Q, Cheng X. AJUBA increases the cisplatin resistance through hippo pathway in cervical cancer. Gene 2017; 644:148-154. [PMID: 29126926 DOI: 10.1016/j.gene.2017.11.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 10/29/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
Though LIM-domain protein AJUBA was identified as a putative oncogene, the function and underlying mechanisms of AJUBA in cervical cancer remain largely unknown. Firstly, AJUBA expression was detected via real-time quantitative PCR in patients' samples. Furthermore, Hela and Siha cells were transfected with AJUBA-overexpressing plasmids, and then exposed to cisplatin, the apoptosis was measured by cytometry assay. In addition, the expression of YAP and TAZ was disclosed through western blot assay. Our results revealed that AJUBA expression was significantly higher in the cervical cancer patients resistant to cisplatin treatment compared with cervical cancer patients sensitive to cisplatin treatment. In addition, overall survival time was significantly shorter in the cervical cancer patients with high AJUBA expression compare with those with low AJUBA expression using kaplan-meier analysis. Hela and Siha cells transfected with AJUBA-expressing plasmids exposed to cisplatin treatment had higher survival rate compared with the cells transfected with empty vector control. Mechanistic studies revealed the AJUBA upregulated the downstream targets YAP and TAZ. These results suggest that high AJUBA level enhances cervical cancer cells drug resistance to cisplatin, also associates with decreased patient survival times.
Collapse
Affiliation(s)
- Lihong Bi
- Department of Gynecology, PKUCare Luzhong Hospital, Zibo, Shandong, China
| | - Feng Ma
- Department of Oncology, PKUCare Luzhong Hospital, Zibo, Shandong, China.
| | - Rui Tian
- Department of Gynecology, PKUCare Luzhong Hospital, Zibo, Shandong, China
| | - Yanli Zhou
- Department of Pharmacy, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Weiguang Lan
- Department of Oncology, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Quanmao Song
- Department of Oncology, PKUCare Luzhong Hospital, Zibo, Shandong, China
| | - Xiankui Cheng
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
20
|
Adamik J, Jin S, Sun Q, Zhang P, Weiss KR, Anderson JL, Silbermann R, Roodman GD, Galson DL. EZH2 or HDAC1 Inhibition Reverses Multiple Myeloma-Induced Epigenetic Suppression of Osteoblast Differentiation. Mol Cancer Res 2017; 15:405-417. [PMID: 28119431 DOI: 10.1158/1541-7786.mcr-16-0242-t] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 01/12/2023]
Abstract
In multiple myeloma, osteolytic lesions rarely heal because of persistent suppressed osteoblast differentiation resulting in a high fracture risk. Herein, chromatin immunoprecipitation analyses reveal that multiple myeloma cells induce repressive epigenetic histone changes at the Runx2 locus that prevent osteoblast differentiation. The most pronounced multiple myeloma-induced changes were at the Runx2-P1 promoter, converting it from a poised bivalent state to a repressed state. Previously, it was observed that multiple myeloma induces the transcription repressor GFI1 in osteoblast precursors, which correlates with decreased Runx2 expression, thus prompting detailed characterization of the multiple myeloma and TNFα-dependent GFI1 response element within the Runx2-P1 promoter. Further analyses reveal that multiple myeloma-induced GFI1 binding to Runx2 in osteoblast precursors and recruitment of the histone modifiers HDAC1, LSD1, and EZH2 is required to establish and maintain Runx2 repression in osteogenic conditions. These GFI1-mediated repressive chromatin changes persist even after removal of multiple myeloma. Ectopic GFI1 is sufficient to bind to Runx2, recruit HDAC1 and EZH2, increase H3K27me3 on the gene, and prevent osteogenic induction of endogenous Runx2 expression. Gfi1 knockdown in MC4 cells blocked multiple myeloma-induced recruitment of HDAC1 and EZH2 to Runx2, acquisition of repressive chromatin architecture, and suppression of osteoblast differentiation. Importantly, inhibition of EZH2 or HDAC1 activity in pre-osteoblasts after multiple myeloma exposure in vitro or in osteoblast precursors from patients with multiple myeloma reversed the repressive chromatin architecture at Runx2 and rescued osteoblast differentiation.Implications: This study suggests that therapeutically targeting EZH2 or HDAC1 activity may reverse the profound multiple myeloma-induced osteoblast suppression and allow repair of the lytic lesions. Mol Cancer Res; 15(4); 405-17. ©2017 AACR.
Collapse
Affiliation(s)
- Juraj Adamik
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shunqian Jin
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Quanhong Sun
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Peng Zhang
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kurt R Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Cancer Stem Cell Laboratory, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Judith L Anderson
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, Indiana
| | - Rebecca Silbermann
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, Indiana
| | - G David Roodman
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, Indiana. .,Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Deborah L Galson
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. .,McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
21
|
The role of the transcriptional repressor growth factor independent 1 in the formation of myeloid cells. Curr Opin Hematol 2017; 24:32-37. [DOI: 10.1097/moh.0000000000000295] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
22
|
Thambyrajah R, Ucanok D, Jalali M, Hough Y, Wilkinson RN, McMahon K, Moore C, Gering M. A gene trap transposon eliminates haematopoietic expression of zebrafish Gfi1aa, but does not interfere with haematopoiesis. Dev Biol 2016; 417:25-39. [PMID: 27432513 PMCID: PMC5003831 DOI: 10.1016/j.ydbio.2016.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/10/2016] [Accepted: 07/15/2016] [Indexed: 11/02/2022]
Abstract
A transposon-mediated gene trap screen identified the zebrafish line qmc551 that expresses a GFP reporter in primitive erythrocytes and also in haemogenic endothelial cells, which give rise to haematopoietic stem and progenitor cells (HSPCs) that seed sites of larval and adult haematopoiesis. The transposon that mediates this GFP expression is located in intron 1 of the gfi1aa gene, one of three zebrafish paralogs that encode transcriptional repressors homologous to mammalian Gfi1 and Gfi1b proteins. In qmc551 transgenics, GFP expression is under the control of the endogenous gfi1aa promoter, recapitulates early gfi1aa expression and allows live observation of gfi1aa promoter activity. While the transposon integration interferes with the expression of gfi1aa mRNA in haematopoietic cells, homozygous qmc551 fish are viable and fertile, and display normal primitive and definitive haematopoiesis. Retained expression of Gfi1b in primitive erythrocytes and up-regulation of Gfi1ab at the onset of definitive haematopoiesis in homozygous qmc551 carriers, are sufficient to allow normal haematopoiesis. This finding contradicts previously published morpholino data that suggested an essential role for zebrafish Gfi1aa in primitive erythropoiesis.
Collapse
Affiliation(s)
- Roshana Thambyrajah
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Deniz Ucanok
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Maryam Jalali
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Yasmin Hough
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Robert Neil Wilkinson
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK; Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Kathryn McMahon
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Chris Moore
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Martin Gering
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| |
Collapse
|
23
|
Kemler D, Dahley O, Roßwag S, Litfin M, Kassel O. The LIM domain protein nTRIP6 acts as a co-repressor for the transcription factor MEF2C in myoblasts. Sci Rep 2016; 6:27746. [PMID: 27292777 PMCID: PMC4904203 DOI: 10.1038/srep27746] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/23/2016] [Indexed: 12/17/2022] Open
Abstract
The transcription factor Myocyte enhancer factor 2C (MEF2C) plays a key role in the late differentiation of skeletal muscle progenitor cells, the so-called myoblasts. During myoblast differentiation, both MEF2C expression and transcriptional activity are regulated. We have reported that nTRIP6, the nuclear isoform of the focal adhesion LIM domain protein TRIP6, acts as an adaptor transcriptional co-activator for several transcription factors. It interacts with the promoter-bound transcription factors and consequently mediates the recruitment of other co-activators. Based on a described interaction between MEF2C and TRIP6 in a yeast-two-hybrid screen, we hypothesised a co-regulatory function of nTRIP6 for MEF2C. In proliferating myoblasts, nTRIP6 interacted with MEF2C and was recruited together with MEF2C to the MEF2-binding regions of the MEF2C target genes Myom2, Mb, Tnni2 and Des. Silencing nTRIP6 or preventing its interaction with MEF2C increased MEF2C transcriptional activity and increased the expression of these MEF2C target genes. Thus, nTRIP6 acts as a co-repressor for MEF2C. Mechanistically, nTRIP6 mediated the recruitment of the class IIa histone deacetylase HDAC5 to the MEF2C-bound promoters. In conclusion, our results unravel a transcriptional co-repressor function for nTRIP6. This adaptor co-regulator can thus exert either co-activator or co-repressor functions, depending on the transcription factor it interacts with.
Collapse
Affiliation(s)
- Denise Kemler
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - Oliver Dahley
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - Sven Roßwag
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - Margarethe Litfin
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - Olivier Kassel
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| |
Collapse
|
24
|
Yasuoka T, Kuwahara M, Yamada T, Maruyama S, Suzuki J, Taniguchi M, Yasukawa M, Yamashita M. The Transcriptional Repressor Gfi1 Plays a Critical Role in the Development of NKT1- and NKT2-Type iNKT Cells. PLoS One 2016; 11:e0157395. [PMID: 27284976 PMCID: PMC4902269 DOI: 10.1371/journal.pone.0157395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/27/2016] [Indexed: 01/09/2023] Open
Abstract
Gfi1 plays an important role in the development and maintenance of many hematopoietic linage cells. However, the impact of Gfi1-deficiency on the iNKT cell differentiation remains unclear. We herein demonstrate a critical role of Gfi1 in regulating the development of iNKT cell subsets. In the thymus of T cell-specific Gfi1-deficient mice, iNKT cells normally developed up to stage 2, while the number of stage 3 NK1.1pos iNKT cells was significantly reduced. Furthermore, CD4pos iNKT cells were selectively reduced in the peripheral organs of T cell-specific Gfi1-deficient mice. The α-GalCer-dependent production of IFN-γand Th2 cytokines, but not IL-17A, was severely reduced in T cell-specific Gfi1-deficient mice. In addition, a reduction of the α-GalCer-induced anti-tumor activity was observed in Gfi1-deficient mice. These findings demonstrate the important role of Gfi1 in regulating the development and function of NKT1- and NKT2-type iNKT cell subsets.
Collapse
Affiliation(s)
- Toshiaki Yasuoka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime, Japan
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime, Japan
| | - Makoto Kuwahara
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime, Japan
- Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime, Japan
- Division of Immune Regulation, Department of Proteo-Inovation, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Takeshi Yamada
- Department of Infection and Host Defenses, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime, Japan
| | - Saho Maruyama
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime, Japan
| | - Junpei Suzuki
- Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime, Japan
- Department of Hematology, Clinical Immunology and Infectious Diseases, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime, Japan
| | - Masaru Taniguchi
- Laboratory of Immune Regulation, RIKEN Center for Integrative Medical Sciences, 1-7-22 suehiro-cho, Tsurumi-ku, Yokohama, Japan
| | - Masaki Yasukawa
- Department of Hematology, Clinical Immunology and Infectious Diseases, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime, Japan
| | - Masakatsu Yamashita
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime, Japan
- Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime, Japan
- Division of Immune Regulation, Department of Proteo-Inovation, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
- * E-mail:
| |
Collapse
|
25
|
Suzuki J, Maruyama S, Tamauchi H, Kuwahara M, Horiuchi M, Mizuki M, Ochi M, Sawasaki T, Zhu J, Yasukawa M, Yamashita M. Gfi1, a transcriptional repressor, inhibits the induction of the T helper type 1 programme in activated CD4 T cells. Immunology 2016; 147:476-87. [PMID: 26749286 DOI: 10.1111/imm.12580] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/13/2015] [Accepted: 01/03/2016] [Indexed: 12/24/2022] Open
Abstract
A transcriptional repressor Gfi1 promotes T helper type 2 (Th2) cell development and inhibits Th17 and inducible regulatory T-cell differentiation. However, the role of Gfi1 in regulating Th1 cell differentiation and the Th1-type immune response remains to be investigated. We herein demonstrate that Gfi1 inhibits the induction of the Th1 programme in activated CD4 T cells. The activated Gfi1-deficient CD4 T cells spontaneously develop into Th1 cells in an interleukin-12- and interferon-γ-independent manner. The increase of Th1-type immune responses was confirmed in vivo in Gfi1-deficient mice using a murine model of nickel allergy and delayed-type hypersensitivity (DTH). The expression levels of Th1-related transcription factors were found to increase in Gfi1-deficient activated CD4 T cells. Tbx21, Eomes and Runx2 were identified as possible direct targets of Gfi1. Gfi1 binds to the Tbx21, Eomes and Runx2 gene loci and reduces the histone H3K4 methylation levels in part by modulating Lsd1 recruitment. Together, these findings demonstrate a novel regulatory role of Gfi1 in the regulation of the Th1-type immune response.
Collapse
Affiliation(s)
- Junpei Suzuki
- Department of Haematology, Clinical Immunology and Infectious Diseases, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan.,Department of Translational Immunology, Translational Research Center, Ehime University Hospital, Toon, Ehime, Japan
| | - Saho Maruyama
- Department of Immunology, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
| | - Hidekazu Tamauchi
- Division of Fundamental Medical Technology, Department of Medical Technology, Ehime Prefectural University of Health Science, Iyo-gun, Ehime, Japan
| | - Makoto Kuwahara
- Department of Translational Immunology, Translational Research Center, Ehime University Hospital, Toon, Ehime, Japan.,Department of Immunology, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan.,Division of Immune Regulation, Department of Proteo-Inovation, Proteo-Science Centre, Ehime University, Matsuyama, Ehime, Japan
| | - Mika Horiuchi
- Department of Immunology, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
| | - Masumi Mizuki
- Department of Immunology, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
| | - Mizuki Ochi
- Department of Immunology, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan.,Division of Cell-Free Sciences, Department of Proteo-Research, Proteo-Science Centre, Ehime University, Matsuyama, Ehime, Japan
| | - Tatsuya Sawasaki
- Division of Cell-Free Sciences, Department of Proteo-Research, Proteo-Science Centre, Ehime University, Matsuyama, Ehime, Japan
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Unit, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Masaki Yasukawa
- Department of Haematology, Clinical Immunology and Infectious Diseases, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
| | - Masakatsu Yamashita
- Department of Translational Immunology, Translational Research Center, Ehime University Hospital, Toon, Ehime, Japan.,Department of Immunology, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan.,Division of Immune Regulation, Department of Proteo-Inovation, Proteo-Science Centre, Ehime University, Matsuyama, Ehime, Japan
| |
Collapse
|
26
|
GFI1 as a novel prognostic and therapeutic factor for AML/MDS. Leukemia 2016; 30:1237-45. [DOI: 10.1038/leu.2016.11] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 01/08/2016] [Accepted: 01/25/2016] [Indexed: 12/17/2022]
|
27
|
Genetic and Epigenetic Mechanisms That Maintain Hematopoietic Stem Cell Function. Stem Cells Int 2015; 2016:5178965. [PMID: 26798358 PMCID: PMC4699043 DOI: 10.1155/2016/5178965] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/03/2015] [Accepted: 09/09/2015] [Indexed: 01/15/2023] Open
Abstract
All hematopoiesis cells develop from multipotent progenitor cells. Hematopoietic stem cells (HSC) have the ability to develop into all blood lineages but also maintain their stemness. Different molecular mechanisms have been identified that are crucial for regulating quiescence and self-renewal to maintain the stem cell pool and for inducing proliferation and lineage differentiation. The stem cell niche provides the microenvironment to keep HSC in a quiescent state. Furthermore, several transcription factors and epigenetic modifiers are involved in this process. These create modifications that regulate the cell fate in a more or less reversible and dynamic way and contribute to HSC homeostasis. In addition, HSC respond in a unique way to DNA damage. These mechanisms also contribute to the regulation of HSC function and are essential to ensure viability after DNA damage. How HSC maintain their quiescent stage during the entire life is still matter of ongoing research. Here we will focus on the molecular mechanisms that regulate HSC function.
Collapse
|
28
|
From cytopenia to leukemia: the role of Gfi1 and Gfi1b in blood formation. Blood 2015; 126:2561-9. [PMID: 26447191 DOI: 10.1182/blood-2015-06-655043] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/06/2015] [Indexed: 12/24/2022] Open
Abstract
The DNA-binding zinc finger transcription factors Gfi1 and Gfi1b were discovered more than 20 years ago and are recognized today as major regulators of both early hematopoiesis and hematopoietic stem cells. Both proteins function as transcriptional repressors by recruiting histone-modifying enzymes to promoters and enhancers of target genes. The establishment of Gfi1 and Gfi1b reporter mice made it possible to visualize their cell type-specific expression and to understand their function in hematopoietic lineages. We now know that Gfi1 is primarily important in myeloid and lymphoid differentiation, whereas Gfi1b is crucial for the generation of red blood cells and platelets. Several rare hematologic diseases are associated with acquired or inheritable mutations in the GFI1 and GFI1B genes. Certain patients with severe congenital neutropenia carry mutations in the GFI1 gene that lead to the disruption of the C-terminal zinc finger domains. Other mutations have been found in the GFI1B gene in families with inherited bleeding disorders. In addition, the Gfi1 locus is frequently found to be a proviral integration site in retrovirus-induced lymphomagenesis, and new, emerging data suggest a role of Gfi1 in human leukemia and lymphoma, underlining the role of both factors not only in normal hematopoiesis, but also in a wide spectrum of human blood diseases.
Collapse
|
29
|
Fan H, Dong W, Li Q, Zou X, Zhang Y, Wang J, Li S, Liu W, Dong Y, Sun H, Hou Z. Ajuba Preferentially Binds LXRα/RXRγ Heterodimer to Enhance LXR Target Gene Expression in Liver Cells. Mol Endocrinol 2015; 29:1608-18. [PMID: 26389695 DOI: 10.1210/me.2015-1046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The liver X receptors (LXRs) are important regulators of lipid, cholesterol, and glucose homeostasis by transcriptional regulation of many key genes in these processes, and the transcriptional activities of LXRs are finely controlled by cooperating with retinoid X receptors and many other coregulators. Here, we report that the LIM protein Ajuba binds to the hinge and the ligand binding domains of LXRα via its C-terminal tandem LIM motifs and enhances LXR target gene expression in liver cells. Depletion of Ajuba in HepG2 cells and in mouse primary hepatocytes decreases LXR target gene expression, whereas stable expression of Ajuba in HepG2 cells results in increased expression of these genes. Mechanistic investigations found that Ajuba selectively interacts with LXRα/retinoid X receptor-γ heterodimer to form a ternary complex, which displays a higher transactivation activity to LXR target genes. Moreover, Ajuba and LXR mutually affect their DNA binding activity at endogenous target chromatins and the cooperation between Ajuba and LXRα is dependent on the functional LXR response elements located in the target promoters. Together, our studies demonstrate that Ajuba is a novel coactivator for LXRs and may play important role in lipid and glucose metabolism.
Collapse
Affiliation(s)
- Hongyan Fan
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Weibing Dong
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Qi Li
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Xiuqun Zou
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Yihong Zhang
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Jiamin Wang
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Shengxian Li
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Wei Liu
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Ying Dong
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Haipeng Sun
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Zhaoyuan Hou
- Department of Endocrinology (H.F., S.L., W.L., Y.D.), Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025 China; Hongqiao Institute of Medicine (H.F., Q.L., X.Z., Y.Z., J.W., Z.H.), Shanghai Tongren Hospital and Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Department of Pathophysiology (W.D., H.S.), Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation (Z.H.), Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
30
|
The LIM protein Ajuba promotes adipogenesis by enhancing PPARγ and p300/CBP interaction. Cell Death Differ 2015; 23:158-68. [PMID: 26113042 DOI: 10.1038/cdd.2015.83] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 05/18/2015] [Accepted: 05/20/2015] [Indexed: 12/16/2022] Open
Abstract
Adipocytes play a vital role in energy homeostasis and adipogenesis is a hierarchically regulated cellular differentiation process, in which the precursor mesenchymal stem cells are differentiated into mature adipocytes. Here, we report Ajuba is an important regulator of adipocyte differentiation by functioning as an obligate co-activator of PPARγ. Ajuba binds the DNA-binding domain of PPARγ via its preLIM region in a ligand-independent manner. Depletion of Ajuba in 3T3-L1 cells decreases PPARγ target gene expression and results in delayed adipogenic differentiation. Conversely, stable overexpression of Ajuba in 3T3-L1 cells increases PPARγ target gene expression and accelerates adipogenic differentiation. Mechanistic investigations demonstrate that Ajuba recruits p300/CBP via its LIM domain and facilitates p300/CBP binding to PPARγ. Moreover, Ajuba, PPARγ, p300/CBP can cooperatively occupy the PPARγ target promoters and concomitantly increases histone acetylation at these loci. Collectively, these data suggest that Ajuba is a co-activator constitutively associated with PPARγ and may be a potential therapeutic target for PPARγ-mediated metabolic disorders.
Collapse
|
31
|
Abstract
The arrival of multicellularity in evolution facilitated cell-cell signaling in conjunction with adhesion. As the ectodomains of cadherins interact with each other directly in trans (as well as in cis), spanning the plasma membrane and associating with multiple other entities, cadherins enable the transduction of "outside-in" or "inside-out" signals. We focus this review on signals that originate from the larger family of cadherins that are inwardly directed to the nucleus, and thus have roles in gene control or nuclear structure-function. The nature of cadherin complexes varies considerably depending on the type of cadherin and its context, and we will address some of these variables for classical cadherins versus other family members. Substantial but still fragmentary progress has been made in understanding the signaling mediators used by varied cadherin complexes to coordinate the state of cell-cell adhesion with gene expression. Evidence that cadherin intracellular binding partners also localize to the nucleus is a major point of interest. In some models, catenins show reduced binding to cadherin cytoplasmic tails favoring their engagement in gene control. When bound, cadherins may serve as stoichiometric competitors of nuclear signals. Cadherins also directly or indirectly affect numerous signaling pathways (e.g., Wnt, receptor tyrosine kinase, Hippo, NFκB, and JAK/STAT), enabling cell-cell contacts to touch upon multiple biological outcomes in embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Pierre D McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center; Program in Genes & Development, Graduate School in Biomedical Sciences, Houston, Texas, USA.
| | - Meghan T Maher
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Cara J Gottardi
- Cellular and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
32
|
Zhang Y, Wang H, Li Y, Xu D, Lu L. Molecular cloning and expression analysis of the Ajuba gene of grass carp (Ctenopharyngodon idella) involved in cellular response to viral infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 48:164-170. [PMID: 25452047 DOI: 10.1016/j.dci.2014.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/06/2014] [Accepted: 10/06/2014] [Indexed: 06/04/2023]
Abstract
Ajuba belongs to the LIM domain proteins, which are involved in the assembly of the extracellular matrix and, along with associated proteins, regulate target genes that connect the extracellular matrix and the cytoskeleton. In the present study, we characterized the entire cDNA sequence of the Ajuba gene from grass carp (gcAjuba). The gcAjuba cDNA contained an open reading frame (ORF) of 2121 bp encoding a polypeptide of 706 amino acids with an estimated molecular mass of 75.966 kDa and three LIM domains in the C-terminal. The transcriptional level of gcAjuba was significantly up-regulated following the stimulation of virus in vitro. Sub-cellular location of gcAjuba and GCRV-JX01 NS26 proteins did not overlap in the cytoplasm and no direct interaction between gcAjuba and the protein NS26 was detected by co-immunoprecipitation (CO-IP) test in grass carp kidney cells. Based on these results, the gcAjuba is determined to be an immediately inducible gene responding to viral infection and in vivo association of gcAjuba with NS26 could not be confirmed, which has been suggested by yeast two-hybrid assay in previous report.
Collapse
Affiliation(s)
- Yanan Zhang
- Key Laboratory of Freshwater Fishery Germplasm Resources, Ministry of Agriculture of P. R. China, Shanghai Ocean University, Shanghai 201306, China
| | - Hao Wang
- Key Laboratory of Freshwater Fishery Germplasm Resources, Ministry of Agriculture of P. R. China, Shanghai Ocean University, Shanghai 201306, China
| | - Yan Li
- Key Laboratory of Freshwater Fishery Germplasm Resources, Ministry of Agriculture of P. R. China, Shanghai Ocean University, Shanghai 201306, China
| | - Dan Xu
- Key Laboratory of Freshwater Fishery Germplasm Resources, Ministry of Agriculture of P. R. China, Shanghai Ocean University, Shanghai 201306, China
| | - Liqun Lu
- Key Laboratory of Freshwater Fishery Germplasm Resources, Ministry of Agriculture of P. R. China, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
33
|
Lange AW, Sridharan A, Xu Y, Stripp BR, Perl AK, Whitsett JA. Hippo/Yap signaling controls epithelial progenitor cell proliferation and differentiation in the embryonic and adult lung. J Mol Cell Biol 2014; 7:35-47. [PMID: 25480985 DOI: 10.1093/jmcb/mju046] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Hippo/Yap pathway is a well-conserved signaling cascade that regulates cell proliferation and differentiation to control organ size and stem/progenitor cell behavior. Following airway injury, Yap was dynamically regulated in regenerating airway epithelial cells. To determine the role of Hippo signaling in the lung, the mammalian Hippo kinases, Mst1 and Mst2, were deleted in epithelial cells of the embryonic and mature mouse lung. Mst1/2 deletion in the fetal lung enhanced proliferation and inhibited sacculation and epithelial cell differentiation. The transcriptional inhibition of cell proliferation and activation of differentiation during normal perinatal lung maturation were inversely regulated following embryonic Mst1/2 deletion. Ablation of Mst1/2 from bronchiolar epithelial cells in the adult lung caused airway hyperplasia and altered differentiation. Inhibitory Yap phosphorylation was decreased and Yap nuclear localization and transcriptional targets were increased after Mst1/2 deletion, consistent with canonical Hippo/Yap signaling. YAP potentiated cell proliferation and inhibited differentiation of human bronchial epithelial cells in vitro. Loss of Mst1/2 and expression of YAP regulated transcriptional targets controlling cell proliferation and differentiation, including Ajuba LIM protein. Ajuba was required for the effects of YAP on cell proliferation in vitro. Hippo/Yap signaling regulates Ajuba and controls proliferation and differentiation of lung epithelial progenitor cells.
Collapse
Affiliation(s)
- Alexander W Lange
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | - Anusha Sridharan
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | - Yan Xu
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | | | - Anne-Karina Perl
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | - Jeffrey A Whitsett
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| |
Collapse
|
34
|
Sourvinos G, Morou A, Sanidas I, Codruta I, Ezell SA, Doxaki C, Kampranis SC, Kottakis F, Tsichlis PN. The downregulation of GFI1 by the EZH2-NDY1/KDM2B-JARID2 axis and by human cytomegalovirus (HCMV) associated factors allows the activation of the HCMV major IE promoter and the transition to productive infection. PLoS Pathog 2014; 10:e1004136. [PMID: 24830456 PMCID: PMC4022736 DOI: 10.1371/journal.ppat.1004136] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 04/07/2014] [Indexed: 12/26/2022] Open
Abstract
Earlier studies had suggested that epigenetic mechanisms play an important role in the control of human cytomegalovirus (HCMV) infection. Here we show that productive HCMV infection is indeed under the control of histone H3K27 trimethylation. The histone H3K27 methyltransferase EZH2, and its regulators JARID2 and NDY1/KDM2B repress GFI1, a transcriptional repressor of the major immediate-early promoter (MIEP) of HCMV. Knocking down EZH2, NDY1/KDM2B or JARID2 relieves the repression and results in the upregulation of GFI1. During infection, the incoming HCMV rapidly downregulates the GFI1 mRNA and protein in both wild-type cells and in cells in which EZH2, NDY1/KDM2B or JARID2 were knocked down. However, since the pre-infection levels of GFI1 in the latter cells are significantly higher, the virus fails to downregulate it to levels permissive for MIEP activation and viral infection. Following the EZH2-NDY1/KDM2B-JARID2-independent downregulation of GFI1 in the early stages of infection, the virus also initiates an EZH2-NDY1/ΚDM2Β-JARID2-dependent program that represses GFI1 throughout the infection cycle. The EZH2 knockdown also delays histone H3K27 trimethylation in the immediate early region of HCMV, which is accompanied by a drop in H3K4 trimethylation that may contribute to the shEZH2-mediated repression of the major immediate early HCMV promoter. These data show that HCMV uses multiple mechanisms to allow the activation of the HCMV MIEP and to prevent cellular mechanisms from blocking the HCMV replication program.
Collapse
Affiliation(s)
- George Sourvinos
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- Laboratory of Virology, Medical School, University of Crete, Heraklion, Crete, Greece
- * E-mail: (GS); (PNT)
| | - Antigoni Morou
- Laboratory of Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Ioannis Sanidas
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Ignea Codruta
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Scott A. Ezell
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Christina Doxaki
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Sotirios C. Kampranis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Filippos Kottakis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Philip N. Tsichlis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- * E-mail: (GS); (PNT)
| |
Collapse
|
35
|
Vásquez-Limeta A, Wagstaff KM, Ortega A, Crouch DH, Jans DA, Cisneros B. Nuclear import of β-dystroglycan is facilitated by ezrin-mediated cytoskeleton reorganization. PLoS One 2014; 9:e90629. [PMID: 24599031 PMCID: PMC3944073 DOI: 10.1371/journal.pone.0090629] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 02/04/2014] [Indexed: 12/12/2022] Open
Abstract
The β-dystroglycan (β-DG) protein has the ability to target to multiple sites in eukaryotic cells, being a member of diverse protein assemblies including the transmembranal dystrophin-associated complex, and a nuclear envelope-localised complex that contains emerin and lamins A/C and B1. We noted that the importin α2/β1-recognised nuclear localization signal (NLS) of β-DG is also a binding site for the cytoskeletal-interacting protein ezrin, and set out to determine whether ezrin binding might modulate β-DG nuclear translocation for the first time. Unexpectedly, we found that ezrin enhances rather than inhibits β-DG nuclear translocation in C2C12 myoblasts. Both overexpression of a phosphomimetic activated ezrin variant (Ez-T567D) and activation of endogenous ezrin through stimulation of the Rho pathway resulted in both formation of actin-rich surface protrusions and significantly increased nuclear translocation of β-DG as shown by quantitative microscopy and subcellular fractionation/Western analysis. In contrast, overexpression of a nonphosphorylatable inactive ezrin variant (Ez-T567A) or inhibition of Rho signaling, decreased nuclear translocation of β-DG concomitant with a lack of cell surface protrusions. Further, a role for the actin cytoskeleton in ezrin enhancement of β-DG nuclear translocation was implicated by the observation that an ezrin variant lacking its actin-binding domain failed to enhance nuclear translocation of β-DG, while disruption of the actin cytoskeleton led to a reduction in β-DG nuclear localization. Finally, we show that ezrin-mediated cytoskeletal reorganization enhances nuclear translocation of the cytoplasmic but not the transmembranal fraction of β-DG. This is the first study showing that cytoskeleton reorganization can modulate nuclear translocation of β-DG, with the implication that β-DG can respond to cytoskeleton-driven changes in cell morphology by translocating from the cytoplasm to the nucleus to orchestrate nuclear processes in response to the functional requirements of the cell.
Collapse
Affiliation(s)
- Alejandra Vásquez-Limeta
- Departamento de Genética y Biología Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México D.F., Mexico
| | - Kylie M. Wagstaff
- Nuclear Signalling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Arturo Ortega
- Departamento de Genética y Biología Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México D.F., Mexico
| | - Dorothy H. Crouch
- School of Dentistry, University of Dundee, Dundee, Scotland, United Kingdom
| | - David A. Jans
- Nuclear Signalling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- * E-mail: (BC); david.jans@ monash.edu (DAJ)
| | - Bulmaro Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México D.F., Mexico
- * E-mail: (BC); david.jans@ monash.edu (DAJ)
| |
Collapse
|
36
|
A novel small compound SH-2251 suppresses Th2 cell-dependent airway inflammation through selective modulation of chromatin status at the Il5 gene locus. PLoS One 2013; 8:e61785. [PMID: 23613936 PMCID: PMC3628909 DOI: 10.1371/journal.pone.0061785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 03/14/2013] [Indexed: 12/18/2022] Open
Abstract
IL-5 is a key cytokine that plays an important role in the development of pathological conditions in allergic inflammation. Identifying strategies to inhibit IL-5 production is important in order to establish new therapies for treating allergic inflammation. We found that SH-2251, a novel thioamide-related small compound, selectively inhibits the differentiation of IL-5-producing Th2 cells. SH-2251 inhibited the induction of active histone marks at the Il5 gene locus during Th2 cell differentiation. The recruitment of RNA polymerase II, and following expression of the Th2 cell-specific intergenic transcripts around the Il5 gene locus was also inhibited. Furthermore, Th2 cell-dependent airway inflammation in mice was suppressed by the oral administration of SH-2251. Gfi1, a transcriptional repressor, was identified as a downstream target molecule of SH-2251 using a DNA microarray analysis. The Gfi1 expression dramatically decreased in SH-2251-treated Th2 cells, and the SH-2251-mediated inhibition of IL-5-producing Th2 cell differentiation was restored by transduction of Gfi1. Therefore, our study unearthed SH-2251 as a novel therapeutic candidate for allergic inflammation that selectively inhibits active histone marks at the Il5 gene locus.
Collapse
|
37
|
Potential roles for Gfi1 in the pathogenesis and proliferation of glioma. Med Hypotheses 2013; 80:629-32. [PMID: 23466061 DOI: 10.1016/j.mehy.2013.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 01/27/2013] [Accepted: 02/07/2013] [Indexed: 11/21/2022]
Abstract
Glioblastoma multiforme (GBM) is a major form of adult brain tumour with relatively poor prognosis and high mortality. Temozolomide (TMZ)-based chemotherapy following neurosurgery and radiotherapy has been suggested as the first line of treatment and is proven to effectively prolong overall survival and enhance patient quality of life. However, not all patients benefit from this treatment because of drug resistance. Even patients with TMZ-sensitive GBM may become resistant, which is partly due to the restoration of activity of the DNA repair enzyme O(6)-methylguanine-DNA-methyltransferase (MGMT); thus, patients cannot evade eventual tumour recurrence. The cellular activity of MGMT is the most important determinant of TMZ-resistance. However, some patients with a low level of activated MGMT are also TMZ-resistant. The aberrant expression of HOXA9, one of the 39 class I homeobox genes, is a marker of poor prognosis, and its level gradually increases with histologic malignant progression, shorter time to overall survival (OS) and free progression survival (FPS) in glioma patients, which further supports an oncogenic role for HOXA9 in gliomas. The HOXA9-PI3K signalling pathway is an important mechanism in GBM that is independent of MGMT promoter methylation status. The DNA binding sites of growth factor independent-1 (Gfi1) can overlap with the HOXA9 promoter through the "AATC" versus "GATT" core sequence. The competition for this binding site inhibits the expression of HOXA9 and induces different transcriptional outcomes, which suggests a new direction for investigation of the mechanism underlying targeted therapy of malignant gliomas.
Collapse
|
38
|
Chiang C, Ayyanathan K. Snail/Gfi-1 (SNAG) family zinc finger proteins in transcription regulation, chromatin dynamics, cell signaling, development, and disease. Cytokine Growth Factor Rev 2012; 24:123-31. [PMID: 23102646 DOI: 10.1016/j.cytogfr.2012.09.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 09/24/2012] [Indexed: 12/12/2022]
Abstract
The Snail/Gfi-1 (SNAG) family of zinc finger proteins is a group of transcriptional repressors that have been intensively studied in mammals. SNAG family members are similarly structured with an N-terminal SNAG repression domain and a C-terminal zinc finger DNA binding domain, however, the spectrum of target genes they regulate and the ranges of biological functions they govern vary widely between them. They play active roles in transcriptional regulation, formation of repressive chromatin structure, cellular signaling and developmental processes. They can also result in disease states due to deregulation. We have performed a thorough investigation of the relevant literature and present a comprehensive mini-review. Based on the available information, we also propose a mechanism by which SNAG family members may function.
Collapse
Affiliation(s)
- Cindy Chiang
- Department of Biological Sciences, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA
| | | |
Collapse
|
39
|
Lancrin C, Mazan M, Stefanska M, Patel R, Lichtinger M, Costa G, Vargel O, Wilson NK, Möröy T, Bonifer C, Göttgens B, Kouskoff V, Lacaud G. GFI1 and GFI1B control the loss of endothelial identity of hemogenic endothelium during hematopoietic commitment. Blood 2012; 120:314-22. [PMID: 22668850 DOI: 10.1182/blood-2011-10-386094] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent studies have established that during embryonic development, hematopoietic progenitors and stem cells are generated from hemogenic endothelium precursors through a process termed endothelial to hematopoietic transition (EHT). The transcription factor RUNX1 is essential for this process, but its main downstream effectors remain largely unknown. Here, we report the identification of Gfi1 and Gfi1b as direct targets of RUNX1 and critical regulators of EHT. GFI1 and GFI1B are able to trigger, in the absence of RUNX1, the down-regulation of endothelial markers and the formation of round cells, a morphologic change characteristic of EHT. Conversely, blood progenitors in Gfi1- and Gfi1b-deficient embryos maintain the expression of endothelial genes. Moreover, those cells are not released from the yolk sac and disseminated into embryonic tissues. Taken together, our findings demonstrate a critical and specific role of the GFI1 transcription factors in the first steps of the process leading to the generation of hematopoietic progenitors from hemogenic endothelium.
Collapse
Affiliation(s)
- Christophe Lancrin
- Cancer Research UK Stem Cell Biology Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Vachon CM, Li J, Scott CG, Hall P, Czene K, Wang X, Liu J, Fredericksen ZS, Rider DN, Wu FF, Olson JE, Cunningham JM, Stevens KN, Sellers TA, Pankratz SV, Couch FJ. No evidence for association of inherited variation in genes involved in mitosis and percent mammographic density. Breast Cancer Res 2012; 14:R7. [PMID: 22226020 PMCID: PMC3496122 DOI: 10.1186/bcr3088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 12/01/2011] [Accepted: 01/07/2012] [Indexed: 01/18/2023] Open
Abstract
Introduction Increased mammographic breast density is one of the strongest risk factors for breast cancer. While two-thirds of the variation in mammographic density appears to be genetically influenced, few variants have been identified. We examined the association of inherited variation in genes from pathways that mediate cell division with percent mammographic density (PMD) adjusted for age, body mass index (BMI) and postmenopausal hormones, in two studies of healthy postmenopausal women. Methods We investigated 2,058 single nucleotide polymorphisms (SNPs) in 378 genes involved in regulation of mitosis for associations with adjusted PMD among 484 unaffected postmenopausal controls (without breast cancer) from the Mayo Clinic Breast Cancer Study (MCBCS) and replicated the findings in postmenopausal controls (n = 726) from the Singapore and Sweden Breast Cancer Study (SASBAC) study. PMD was assessed in both studies by a computer-thresholding method (Cumulus) and linear regression approaches were used to assess the association of SNPs and PMD, adjusted for age, BMI and postmenopausal hormones. A P-value threshold of 4.2 × 10-5 based on a Bonferroni correction of effective number of independent tests was used for statistical significance. Further, a pathway-level analysis was conducted of all 378 genes using the self-contained gene-set analysis method GLOSSI. Results A variant in PRPF4, rs10733604, was significantly associated with adjusted PMD in the MCBCS (P = 2.7 × 10-7), otherwise, no single SNP was associated with PMD. Additionally, the pathway analysis provided no evidence of enrichment in the number of associations observed between SNPs in the mitotic genes and PMD (P = 0.60). We evaluated rs10733604 (PRPF4), and 73 other SNPs at P < 0.05 from 51 genes in the SASBAC study. There was no evidence of an association of rs10733604 (PRPF4) with adjusted PMD in SASBAC (P = 0.23). There were, however, consistent associations (P < 0.05) of variants at the putative locus, LOC375190, Aurora B kinase (AURKB), and Mini-chromosome maintenance complex component 3 (MCM3) with adjusted PMD, although these were not statistically significant. Conclusions Our findings do not support a role of inherited variation in genes involved in regulation of cell division and adjusted percent mammographic density in postmenopausal women.
Collapse
Affiliation(s)
- Celine M Vachon
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Nola S, Daigaku R, Smolarczyk K, Carstens M, Martin-Martin B, Longmore G, Bailly M, Braga VMM. Ajuba is required for Rac activation and maintenance of E-cadherin adhesion. ACTA ACUST UNITED AC 2011; 195:855-71. [PMID: 22105346 PMCID: PMC3257575 DOI: 10.1083/jcb.201107162] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A Rac–PAK1–Ajuba feedback loop stabilizes cadherin complexes via coordination of spatiotemporal signaling with actin remodeling at cell–cell contacts. Maintenance of stable E-cadherin–dependent adhesion is essential for epithelial function. The small GTPase Rac is activated by initial cadherin clustering, but the precise mechanisms underlying Rac-dependent junction stabilization are not well understood. Ajuba, a LIM domain protein, colocalizes with cadherins, yet Ajuba function at junctions is unknown. We show that, in Ajuba-depleted cells, Rac activation and actin accumulation at cadherin receptors was impaired, and junctions did not sustain mechanical stress. The Rac effector PAK1 was also transiently activated upon cell–cell adhesion and directly phosphorylated Ajuba (Thr172). Interestingly, similar to Ajuba depletion, blocking PAK1 activation perturbed junction maintenance and actin recruitment. Expression of phosphomimetic Ajuba rescued the effects of PAK1 inhibition. Ajuba bound directly to Rac·GDP or Rac·GTP, but phosphorylated Ajuba interacted preferentially with active Rac. Rather than facilitating Rac recruitment to junctions, Ajuba modulated Rac dynamics at contacts depending on its phosphorylation status. Thus, a Rac–PAK1–Ajuba feedback loop integrates spatiotemporal signaling with actin remodeling at cell–cell contacts and stabilizes preassembled cadherin complexes.
Collapse
Affiliation(s)
- Sébastien Nola
- Molecular Medicine, National Heart and Lung Institute, Imperial College London, London SW7 2AZ, England, UK
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Gfi1 expressed in bone marrow stromal cells is a novel osteoblast suppressor in patients with multiple myeloma bone disease. Blood 2011; 118:6871-80. [PMID: 22042697 DOI: 10.1182/blood-2011-04-346775] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Protracted inhibition of osteoblast (OB) differentiation characterizes multiple myeloma (MM) bone disease and persists even when patients are in long-term remission. However, the underlying pathophysiology for this prolonged OB suppression is unknown. Therefore, we developed a mouse MM model in which the bone marrow stromal cells (BMSCs) remained unresponsive to OB differentiation signals after removal of MM cells. We found that BMSCs from both MM-bearing mice and MM patients had increased levels of the transcriptional repressor Gfi1 compared with controls and that Gfi1 was a novel transcriptional repressor of the critical OB transcription factor Runx2. Trichostatin-A blocked the effects of Gfi1, suggesting that it induces epigenetic changes in the Runx2 promoter. MM-BMSC cell-cell contact was not required for MM cells to increase Gfi1 and repress Runx2 levels in MC-4 before OBs or naive primary BMSCs, and Gfi1 induction was blocked by anti-TNF-α and anti-IL-7 antibodies. Importantly, BMSCs isolated from Gfi1(-/-) mice were significantly resistant to MM-induced OB suppression. Strikingly, siRNA knockdown of Gfi1 in BMSCs from MM patients significantly restored expression of Runx2 and OB differentiation markers. Thus, Gfi1 may have an important role in prolonged MM-induced OB suppression and provide a new therapeutic target for MM bone disease.
Collapse
|
43
|
Möröy T, Khandanpour C. Growth factor independence 1 (Gfi1) as a regulator of lymphocyte development and activation. Semin Immunol 2011; 23:368-78. [PMID: 21920773 DOI: 10.1016/j.smim.2011.08.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 08/19/2011] [Indexed: 10/17/2022]
Abstract
T- and B-lymphocytes are important elements in the immune defense repertoire of higher organisms. The development and function of lymphoid cells is regulated at many levels one being the control of gene expression by transcription factors. The zinc finger transcriptional repressor Gfi1 has emerged as a factor that is critically implicated in the commitment of precursor cells for the lymphoid lineage. In addition, Gfi1 controls distinct stages of early T- or B-lymphoid development and is also critical for their maturation, activation and effector function. From many years of work, a picture emerges in which Gfi1 is part of a complicated, but well orchestrated network of interdependent regulators, most of which impinge on lymphoid development and activation by transcriptional regulation. Biochemical studies show that Gfi1 is part of a large DNA binding multi-protein complex that enables histone modifications, but may also control alternative pre mRNA splicing. Many insights into the biological role of Gfi1 have been gained through the study of gene deficient mice that have defects in B- and T-cell differentiation, in T-cell selection and polarization processes and in the response of mature B- and T-cells towards antigen. Most importantly, the defects seen in Gfi1 deficient mice also point to roles of Gfi1 in diseases of the immune system that involve auto-immune responses and acute lymphoid leukemia and lymphoma.
Collapse
Affiliation(s)
- Tarik Möröy
- Institut de recherches cliniques de Montréal - IRCM, 110 Avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada.
| | | |
Collapse
|
44
|
Heyd F, Chen R, Afshar K, Saba I, Lazure C, Fiolka K, Möröy T. The p150 subunit of the histone chaperone Caf-1 interacts with the transcriptional repressor Gfi1. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1809:255-61. [PMID: 21570500 DOI: 10.1016/j.bbagrm.2011.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 04/07/2011] [Accepted: 04/26/2011] [Indexed: 11/29/2022]
Abstract
Modification of histones is critically involved in regulating chromatin structure and gene expression. The zinc finger protein Gfi1 silences transcription by recruiting a complex of histone modifying enzymes such as LSD-1/CoRest and HDAC-1 to target gene promoters. Here we present evidence that Gfi1 forms a complex with the p150 subunit of the histone chaperone chromatin assembly factor-1 (Caf-1). Gfi1 and p150 interact at endogenous expression levels and co-localize in distinct sub-nuclear structures. We show that p150 enhances Gfi1-mediated transcriptional repression and that it occupies Gfi1 target gene promoters in transfected cells and primary murine T cells only in the presence of Gfi1. Finally, size exclusion chromatography shows a fraction of p150 to coelute with Gfi1, LSD-1 and HDAC-1 and thus provides evidence that p150 is part of the Gfi1 repression complex. Since p150 binds directly to histones H3 and H4, our findings suggest that p150 may link the DNA-bound Gfi1 repressor complex to histones enabling modifications required for transcriptional silencing.
Collapse
Affiliation(s)
- Florian Heyd
- Institut de recherches cliniques de Montréal (IRCM), H2W 1R7, Montréal, QC, Canada
| | | | | | | | | | | | | |
Collapse
|
45
|
Kenth G, Puzhko S, Goodyer CG. Human growth hormone receptor gene expression is regulated by Gfi-1/1b and GAGA cis-elements. Mol Cell Endocrinol 2011; 335:135-47. [PMID: 21238539 DOI: 10.1016/j.mce.2011.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Revised: 12/20/2010] [Accepted: 01/07/2011] [Indexed: 12/15/2022]
Abstract
Human growth hormone receptor (hGHR) gene regulation is complex: mRNAs are transcribed from multiple variant (V) 5'UTR exons, several ubiquitously while others only in the postnatal hepatocyte. The liver-specific V1 exon promoter contains Gfi-1/1b repressor sites adjacent to a GAGA box, a GH response element (GHRE) in several mammalian genes. GAGA boxes are also present in the ubiquitously expressing V3 exon promoter. Heterologous sites in bovine, ovine and murine GHR genes suggest conserved roles. GAGA factor stimulated V1 and V3 promoters while Gfi-1/1b repressed basal and GAF-stimulated V1 transcription. HGH treatment of HepG2 cells resulted in a new complex forming with V3 GAGA elements, suggesting a functional GHRE. Data suggest liver-specific V1 transcription is regulated by inhibitory Gfi-1/1b and stimulatory GAGA cis-elements and Gfi-1/1b may control the lack of V1 expression in fetal liver, hepatic tumours and non-hepatic tissues. In addition, hGH may regulate hGHR expression through V3 GAGA boxes.
Collapse
Affiliation(s)
- Gurvinder Kenth
- Department of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
46
|
Gfi1-cells and circuits: unraveling transcriptional networks of development and disease. Curr Opin Hematol 2010; 17:300-7. [PMID: 20571393 DOI: 10.1097/moh.0b013e32833a06f8] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The review will integrate current knowledge of transcriptional circuits whose dysregulation leads to autoimmunity, neutropenia and leukemia. RECENT FINDINGS Growth factor independent-1 (Gfi1) is a transcriptional repressor with essential roles in controlling hematopoietic stem cell biology, myeloid and lymphoid differentiation and lymphocyte effector functions. Recent work has suggested that Gfi1 competes or collaborates with other transcription factors to modulate transcription programs and lineage decisions. SUMMARY Gfi1 is central to several transcriptional circuits whose dysregulation leads to abnormal or malignant hematopoiesis. These functional relationships are conserved from Drosophila development. Such conserved pathways represent central oncogenic or 'gatekeeper' pathways that are pivotal to understanding the process of cellular transformation, and illustrate key targets for clinical intervention.
Collapse
|
47
|
van der Meer LT, Jansen JH, van der Reijden BA. Gfi1 and Gfi1b: key regulators of hematopoiesis. Leukemia 2010; 24:1834-43. [DOI: 10.1038/leu.2010.195] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
48
|
Zinc finger protein Gfi1 controls the endotoxin-mediated Toll-like receptor inflammatory response by antagonizing NF-kappaB p65. Mol Cell Biol 2010; 30:3929-42. [PMID: 20547752 DOI: 10.1128/mcb.00087-10] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Endotoxin (bacterial lipopolysaccharide [LPS]) causes fatal septic shock via the Toll-like receptor 4 (TLR-4) protein present on innate immunity effector cells, which activates nuclear factor kappa B (NF-kappaB), inducing proinflammatory cytokines, including tumor necrosis factor alpha (TNF-alpha). An early step in this process involves nuclear sequestration of the p65-RelA NF-kappaB subunit, enabling transcriptional activation of target inflammatory cytokine genes. Here, we analyzed the role of the nuclear zinc finger protein Gfi1 in the TLR response using primary bone marrow-derived macrophages. We show that upon LPS stimulation, expression of Gfi1 is induced with kinetics similar to those of nuclear translocation of p65 and that Gfi1 interacts with p65 and inhibits p65-mediated transcriptional transactivation by interfering with p65 binding to target gene promoter DNA. Gfi1-deficient macrophages show abnormally high mRNA levels of the TNF-alpha gene and many other p65 target genes and a higher rate of TNF promoter occupancy by p65 than wild-type cells after LPS stimulation, suggesting that Gfi1 functions as an antagonist of NF-kappaB activity at the level of promoter binding. Our findings identify a new function of Gfi1 as a general negative regulator of the endotoxin-initiated innate immune responses, including septic shock and possibly other severe inflammatory diseases.
Collapse
|
49
|
The transcription factor Gfi1 regulates G-CSF signaling and neutrophil development through the Ras activator RasGRP1. Blood 2010; 115:3970-9. [PMID: 20203268 DOI: 10.1182/blood-2009-10-246967] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The transcription factor growth factor independence 1 (Gfi1) and the growth factor granulocyte colony-stimulating factor (G-CSF) are individually essential for neutrophil differentiation from myeloid progenitors. Here, we provide evidence that the functions of Gfi1 and G-CSF are linked in the regulation of granulopoiesis. We report that Gfi1 promotes the expression of Ras guanine nucleotide releasing protein 1 (RasGRP1), an exchange factor that activates Ras, and that RasGRP1 is required for G-CSF signaling through the Ras/mitogen-activated protein/extracellular signal-regulated kinase (MEK/Erk) pathway. Gfi1-null mice have reduced levels of RasGRP1 mRNA and protein in thymus, spleen, and bone marrow, and Gfi1 transduction in myeloid cells promotes RasGRP1 expression. When stimulated with G-CSF, Gfi1-null myeloid cells are selectively defective at activating Erk1/2, but not signal transducer and activator of transcription 1 (STAT1) or STAT3, and fail to differentiate into neutrophils. Expression of RasGRP1 in Gfi1-deficient cells rescues Erk1/2 activation by G-CSF and allows neutrophil maturation by G-CSF. These results uncover a previously unknown function of Gfi1 as a regulator of RasGRP1 and link Gfi1 transcriptional control to G-CSF signaling and regulation of granulopoiesis.
Collapse
|
50
|
LIM protein Ajuba functions as a nuclear receptor corepressor and negatively regulates retinoic acid signaling. Proc Natl Acad Sci U S A 2010; 107:2938-43. [PMID: 20133701 DOI: 10.1073/pnas.0908656107] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Corepressors play an essential role in nuclear receptor-mediated transcriptional repression. In general, corepressors directly bind to nuclear receptors via CoRNR boxes (L/I-X-X-I/V-I) in the absence of ligand and appear to act as scaffolds to further recruit chromatin remodeling complexes to specific target genes. Here, we describe the identification of the multiple LIM domain protein Ajuba as a unique corepressor for a subset of nuclear hormone receptors. Ajuba contains functional nuclear-receptor interacting motifs and selectively interacts with retinoic acid receptors (RARs) and rexinoid receptor (RXRs) subtypes in a ligand-dependent manner. Simultaneous mutation of these motifs abolishes RAR binding and concomitantly leads to loss of repression on RARE reporter genes. P19 cells depleted of Ajuba are highly sensitized to all-trans retinoic acid (atRA)-induced transcription and differentiation. In the absence of atRA, Ajuba can be readily found at the RARE control elements of RAR endogenous target genes. Stimulation of cells with atRA results in the dissociation of Ajuba from these regions. Moreover, we observed that coexpression of the known Ajuba binding partner Prmt5 (protein arginine methyltransferase-5) inhibited the Ajuba/RAR interaction. The high-affinity Ajuba-RAR/RXR interaction site overlaps the region responsible for Ajuba/Prmt5 binding, and thus binding appears to be mutually exclusive, providing a potential mechanism for these observations. Identification of Ajuba as a unique corepressor for nuclear receptors sheds new light on mechanisms for nuclear receptor-mediated repression and provides a unique target for developing more effective therapeutics to modulate this important pathway.
Collapse
|