1
|
Wong KW, Zeng Y, Tay E, Teo JHJ, Cipta NO, Hamashima K, Yi Y, Liu H, Warrier T, Le MTN, Ng SC, Li QJ, Li H, Loh YH. Nuclear receptor-SINE B1 network modulates expanded pluripotency in blastoids and blastocysts. Nat Commun 2024; 15:10011. [PMID: 39562549 PMCID: PMC11577042 DOI: 10.1038/s41467-024-54381-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Embryonic stem cells possess the remarkable ability to self-organize into blastocyst-like structures upon induction. These stem cell-based embryo models serve as invaluable platforms for studying embryogenesis and therapeutic developments. Nevertheless, the specific intrinsic regulators that govern this potential for blastoid formation remain unknown. Here we demonstrate an intrinsic program that plays a crucial role in both blastoids and blastocysts across multiple species. We first establish metrics for grading the resemblance of blastoids to mouse blastocysts, and identify the differential activation of gene regulons involved in lineage specification among various blastoid grades. Notably, abrogation of nuclear receptor subfamily 1, group H, member 2 (Nr1h2) drastically reduces blastoid formation. Nr1h2 activation alone is sufficient to rewire conventional ESC into a distinct pluripotency state, enabling them to form blastoids with enhanced implantation capacity in the uterus and contribute to both embryonic and extraembryonic lineages in vivo. Through integrative multi-omics analyses, we uncover the broad regulatory role of Nr1h2 in the transcriptome, chromatin accessibility and epigenome, targeting genes associated with embryonic lineage and the transposable element SINE-B1. The Nr1h2-centred intrinsic program governs and drives the development of both blastoids and early embryos.
Collapse
Grants
- R03 OD038392 NIH HHS
- U19 AG074879 NIA NIH HHS
- R03 OD034496 NIH HHS
- P30 CA015083 NCI NIH HHS
- P30 DK084567 NIDDK NIH HHS
- P50 CA136393 NCI NIH HHS
- National Research Foundation, Singapore (NRF) Investigatorship award [NRFI2018- 02]; National Medical Research Council [NMRC/OFIRG21nov-0088]; Singapore Food Story (SFS) R&D Programme [W22W3D0007]; A*STAR Biomedical Research Council, Central Research Fund, Use-Inspired Basic Research (CRF UIBR); Competitive Research Programme (CRP) [NRF-CRP29-2022-0005]; Industry Alignment Fund - Prepositioning (IAF-PP) [H23J2a0095, H23J2a0097].
- NMRC grant MOH-000937-00 and A*STAR grant C210812003
- M.T.N.L. was supported by the Industry Alignment Fund - Prepositioning (IAF-PP) [H23J2a0097].
- H.L. was supported by grants from the Mayo Clinic Center for Biomedical Discovery, Center for Individualized Medicine, the Mayo Clinic Comprehensive Cancer Center (NIH; P30CA015083), the Mayo Clinic Center for Cell Signaling in Gastroenterology (NIH: P30DK084567), the Mayo Clinic Nutrition Obesity Research Program, the Glenn Foundation for Medical Research, the Eric & Wendy Schmidt Fund for AI Research & Innovation and the National Institutes of Health (NIH; U19AG74879, P50CA136393, R03OD038392).
Collapse
Affiliation(s)
- Ka Wai Wong
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Yingying Zeng
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Republic of Singapore
| | - Edison Tay
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Jia Hao Jackie Teo
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Nadia Omega Cipta
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Kiyofumi Hamashima
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Yao Yi
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Haijun Liu
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Tushar Warrier
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Minh T N Le
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Soon Chye Ng
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Sincere Healthcare Group, Singapore, Republic of Singapore
| | - Qi-Jing Li
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore, 138648, Republic of Singapore
| | - Hu Li
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Yuin-Han Loh
- Cell Fate Engineering and Therapeutics Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
- Department of Biological Sciences, National University of Singapore, Singapore, Republic of Singapore.
- NUS Graduate School's Integrative Sciences and Engineering Programme, National University of Singapore, Singapore, Republic of Singapore.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
2
|
Wu Z, Ma X, Wang J. NCOA3 knockdown delays human embryo development. Heliyon 2024; 10:e37639. [PMID: 39315150 PMCID: PMC11417216 DOI: 10.1016/j.heliyon.2024.e37639] [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: 05/05/2024] [Revised: 08/17/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
Embryonic development is a precisely controlled sequential process influenced by complex external and internal factors; therefore, this process holds paramount significance in the context of in vitro fertilization and embryo transfer (IVF-ET), with internal oocyte and embryo quality being pivotal in determining its success. Nuclear receptor coactivator 3 (NCOA3), a member of the p160 nuclear receptor coactivators family, has been extensively studied in tumorigenesis and reportedly plays a crucial role in maintaining pluripotency in mouse embryonic stem cells (ESCs). However, its functions in human embryo development remain largely unexplored. In this study, we collected human samples, including oocytes, zygotes, and embryos, from patients at the First Affiliated Hospital of Zhengzhou University to investigate whether NCOA3 regulates human embryonic development. To this end, we employed various assays, including immunofluorescence, quantitative real-time PCR (qPCR), microinjection, and RNA sequencing. Our findings suggested that NCOA3 expression level was low in inferior embryos (with >50 % fragmentation), and its presence is closely related to the expression of the pluripotency factor NANOG. Deletion of NCOA3 delays human embryonic development. Single-oocyte RNA sequencing revealed that NCOA3 primarily participates in metabolic alterations in oocytes. In sum, these findings elucidate the pivotal roles of NCOA3 in human embryonic development-NCOA3 deletion compromise the developmental potential of embryos. These mechanistic insights into the role of NCOA3 in human embryonic development not only advances our understanding of the intricate molecular mechanisms involved but also holds potential implications for improving assisted reproductive technologies (ART) and addressing developmental disorders in human embryos.
Collapse
Affiliation(s)
- Zhaoting Wu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Xueshan Ma
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Department of Reproduction and Genetics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, China
| | - Jingyu Wang
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
3
|
Xie Y, Peng X, Li P. MIWE: detecting the critical states of complex biological systems by the mutual information weighted entropy. BMC Bioinformatics 2024; 25:44. [PMID: 38280998 PMCID: PMC10822190 DOI: 10.1186/s12859-024-05667-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/22/2024] [Indexed: 01/29/2024] Open
Abstract
Complex biological systems often undergo sudden qualitative changes during their dynamic evolution. These critical transitions are typically characterized by a catastrophic progression of the system. Identifying the critical point is critical to uncovering the underlying mechanisms of complex biological systems. However, the system may exhibit minimal changes in its state until the critical point is reached, and in the face of high throughput and strong noise data, traditional biomarkers may not be effective in distinguishing the critical state. In this study, we propose a novel approach, mutual information weighted entropy (MIWE), which uses mutual information between genes to build networks and identifies critical states by quantifying molecular dynamic differences at each stage through weighted differential entropy. The method is applied to one numerical simulation dataset and four real datasets, including bulk and single-cell expression datasets. The critical states of the system can be recognized and the robustness of MIWE method is verified by numerical simulation under the influence of different noises. Moreover, we identify two key transcription factors (TFs), CREB1 and CREB3, that regulate downstream signaling genes to coordinate cell fate commitment. The dark genes in the single-cell expression datasets are mined to reveal the potential pathway regulation mechanism.
Collapse
Affiliation(s)
- Yuke Xie
- School of Mathematics and Statistics, Henan University of Science and Technology, Luoyang, 471000, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Xueqing Peng
- School of Mathematics and Statistics, Henan University of Science and Technology, Luoyang, 471000, China
| | - Peiluan Li
- School of Mathematics and Statistics, Henan University of Science and Technology, Luoyang, 471000, China.
| |
Collapse
|
4
|
Hereñú CB, Crespo R. Phytochemicals as estrogen receptor modulators?-a commentary of a network pharmacology study of two commonly employed Chinese herbal medicines in non-small cell lung cancer treatment. Transl Cancer Res 2023; 12:3249-3254. [PMID: 38197069 PMCID: PMC10774059 DOI: 10.21037/tcr-23-1440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/30/2023] [Indexed: 01/11/2024]
Affiliation(s)
- Claudia Beatriz Hereñú
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Rosana Crespo
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| |
Collapse
|
5
|
Varisli L, Dancik GM, Tolan V, Vlahopoulos S. Critical Roles of SRC-3 in the Development and Progression of Breast Cancer, Rendering It a Prospective Clinical Target. Cancers (Basel) 2023; 15:5242. [PMID: 37958417 PMCID: PMC10648290 DOI: 10.3390/cancers15215242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer (BCa) is the most frequently diagnosed malignant tumor in women and is also one of the leading causes of cancer-related death. Most breast tumors are hormone-dependent and estrogen signaling plays a critical role in promoting the survival and malignant behaviors of these cells. Estrogen signaling involves ligand-activated cytoplasmic estrogen receptors that translocate to the nucleus with various co-regulators, such as steroid receptor co-activator (SRC) family members, and bind to the promoters of target genes and regulate their expression. SRC-3 is a member of this family that interacts with, and enhances, the transcriptional activity of the ligand activated estrogen receptor. Although SRC-3 has important roles in normal homeostasis and developmental processes, it has been shown to be amplified and overexpressed in breast cancer and to promote malignancy. The malignancy-promoting potential of SRC-3 is diverse and involves both promoting malignant behavior of tumor cells and creating a tumor microenvironment that has an immunosuppressive phenotype. SRC-3 also inhibits the recruitment of tumor-infiltrating lymphocytes with effector function and promotes stemness. Furthermore, SRC-3 is also involved in the development of resistance to hormone therapy and immunotherapy during breast cancer treatment. The versatility of SRC-3 in promoting breast cancer malignancy in this way makes it a good target, and methodical targeting of SRC-3 probably will be important for the success of breast cancer treatment.
Collapse
Affiliation(s)
- Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey;
| | - Garrett M. Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA;
| | - Veysel Tolan
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey;
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece
| |
Collapse
|
6
|
Shimada MK. Splicing Modulators Are Involved in Human Polyglutamine Diversification via Protein Complexes Shuttling between Nucleus and Cytoplasm. Int J Mol Sci 2023; 24:ijms24119622. [PMID: 37298574 DOI: 10.3390/ijms24119622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Length polymorphisms of polyglutamine (polyQs) in triplet-repeat-disease-causing genes have diversified during primate evolution despite them conferring a risk of human-specific diseases. To explain the evolutionary process of this diversification, there is a need to focus on mechanisms by which rapid evolutionary changes can occur, such as alternative splicing. Proteins that can bind polyQs are known to act as splicing factors and may provide clues about the rapid evolutionary process. PolyQs are also characterized by the formation of intrinsically disordered (ID) regions, so I hypothesized that polyQs are involved in the transportation of various molecules between the nucleus and cytoplasm to regulate mechanisms characteristic of humans such as neural development. To determine target molecules for empirical research to understand the evolutionary change, I explored protein-protein interactions (PPIs) involving the relevant proteins. This study identified pathways related to polyQ binding as hub proteins scattered across various regulatory systems, including regulation via PQBP1, VCP, or CREBBP. Nine ID hub proteins with both nuclear and cytoplasmic localization were found. Functional annotations suggested that ID proteins containing polyQs are involved in regulating transcription and ubiquitination by flexibly changing PPI formation. These findings explain the relationships among splicing complex, polyQ length variations, and modifications in neural development.
Collapse
Affiliation(s)
- Makoto K Shimada
- Center for Medical Science, Fujita Health University, Toyoake 470-1192, Japan
| |
Collapse
|
7
|
Pavani KC, Meese T, Pascottini OB, Guan X, Lin X, Peelman L, Hamacher J, Van Nieuwerburgh F, Deforce D, Boel A, Heindryckx B, Tilleman K, Van Soom A, Gadella BM, Hendrix A, Smits K. Hatching is modulated by microRNA-378a-3p derived from extracellular vesicles secreted by blastocysts. Proc Natl Acad Sci U S A 2022; 119:e2122708119. [PMID: 35298333 PMCID: PMC8944274 DOI: 10.1073/pnas.2122708119] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/04/2022] [Indexed: 12/17/2022] Open
Abstract
SignificanceHatching from the zona pellucida is a prerequisite for embryo implantation and is less likely to occur in vitro for reasons unknown. Extracellular vesicles (EVs) are secreted by the embryo into the culture medium. Yet the role that embryonic EVs and their cargo microRNAs (miRNAs) play in blastocyst hatching has not been elucidated, partially due to the difficulties of isolating them from low amounts of culture medium. Here, we optimized EV-miRNA isolation from medium conditioned by individually cultured bovine embryos and subsequently showed that miR-378a-3p, which was up-regulated in EVs secreted by blastocysts, plays a crucial role in promoting blastocyst hatching. This demonstrates the regulatory effect of miR-378-3p on hatching, which is an established embryo quality parameter linked with implantation.
Collapse
Affiliation(s)
- Krishna Chaitanya Pavani
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, B-9820 Merelbeke, Belgium
- Department for Reproductive Medicine, Ghent University Hospital, 9000 Gent, Belgium
| | - Tim Meese
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Osvaldo Bogado Pascottini
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, B-9820 Merelbeke, Belgium
- Department of Veterinary Sciences, Gamete Research Center, University of Antwerp, 2610 Antwerp, Belgium
| | - XueFeng Guan
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000 Ghent, Belgium
| | - Xiaoyuan Lin
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000 Ghent, Belgium
| | - Luc Peelman
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000 Ghent, Belgium
| | - Joachim Hamacher
- Institute of Crop Science and Resource Conservation, Plant Pathology, Rheinische Friedrich-Wilhelms-University of Bonn, D-53115 Bonn, Germany
| | - Filip Van Nieuwerburgh
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Dieter Deforce
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Annekatrien Boel
- Ghent-Fertility and Stem Cell Team, Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Björn Heindryckx
- Ghent-Fertility and Stem Cell Team, Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Kelly Tilleman
- Department for Reproductive Medicine, Ghent University Hospital, 9000 Gent, Belgium
| | - Ann Van Soom
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, B-9820 Merelbeke, Belgium
| | - Bart M. Gadella
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, B-9000 Ghent, Belgium
- Cancer Research Institute Ghent, B-9000 Ghent, Belgium
| | - Katrien Smits
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, B-9820 Merelbeke, Belgium
| |
Collapse
|
8
|
Huang Y, Duan X, Wang Z, Sun Y, Guan Q, Kang L, Zhang Q, Fang L, Li J, Wong J. An acetylation-enhanced interaction between transcription factor Sox2 and the steroid receptor coactivators facilitates Sox2 transcriptional activity and function. J Biol Chem 2021; 297:101389. [PMID: 34762910 PMCID: PMC8668987 DOI: 10.1016/j.jbc.2021.101389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/02/2022] Open
Abstract
SRY-box 2 (Sox2) is a transcription factor with critical roles in maintaining embryonic stem (ES) cell and adult stem cell functions and in tumorigenesis. However, how Sox2 exerts its transcriptional function remains unclear. Here, we used an in vitro protein–protein interaction assay to discover transcriptional regulators for ES cell core transcription factors (Oct4, Sox2, Klf4, and c-Myc) and identified members of the steroid receptor coactivators (SRCs) as Sox2-specific interacting proteins. The SRC family coactivators have broad roles in transcriptional regulation, but it is unknown whether they also serve as Sox2 coactivators. We demonstrated that these proteins facilitate Sox2 transcriptional activity and act synergistically with p300. Furthermore, we uncovered an acetylation-enhanced interaction between Sox2 and SRC-2/3, but not SRC-1, demonstrating it is Sox2 acetylation that promotes the interaction. We identified putative Sox2 acetylation sites required for acetylation-enhanced interaction between Sox2 and SRC-3 and demonstrated that acetylation on these sites contributes to Sox2 transcriptional activity and recruitment of SRC-3. We showed that activation domains 1 and 2 of SRC-3 both display a preferential binding to acetylated Sox2. Finally, functional analyses in mouse ES cells demonstrated that knockdown of SRC-2/3 but not SRC-1 in mouse ES cells significantly downregulates the transcriptional activities of various Sox2 target genes and impairs ES cell stemness. Taken together, we identify specific SRC family proteins as novel Sox2 coactivators and uncover the role of Sox2 acetylation in promoting coactivator recruitment and Sox2 transcriptional function.
Collapse
Affiliation(s)
- Yuanyong Huang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiaoya Duan
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhen Wang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yimei Sun
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qingqing Guan
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Li Kang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qiao Zhang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Lan Fang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiwen Li
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China; Joint Center for Translational Medicine, Fengxian District Central Hospital, Shanghai, China.
| |
Collapse
|
9
|
SRC3 acetylates calmodulin in the mouse brain to regulate synaptic plasticity and fear learning. J Biol Chem 2021; 297:101044. [PMID: 34358562 PMCID: PMC8390517 DOI: 10.1016/j.jbc.2021.101044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/20/2021] [Accepted: 08/02/2021] [Indexed: 11/23/2022] Open
Abstract
Protein acetylation is a reversible posttranslational modification, which is regulated by lysine acetyltransferase (KAT) and lysine deacetyltransferase (KDAC). Although protein acetylation has been shown to regulate synaptic plasticity, this was mainly for histone protein acetylation. The function and regulation of nonhistone protein acetylation in synaptic plasticity and learning remain largely unknown. Calmodulin (CaM), a ubiquitous Ca2+ sensor, plays critical roles in synaptic plasticity such as long-term potentiation (LTP). During LTP induction, activation of NMDA receptor triggers Ca2+ influx, and the Ca2+ binds with CaM and activates calcium/calmodulin-dependent protein kinase IIα (CaMKIIα). In our previous study, we demonstrated that acetylation of CaM was important for synaptic plasticity and fear learning in mice. However, the KAT responsible for CaM acetylation is currently unknown. Here, following an HEK293 cell-based screen of candidate KATs, steroid receptor coactivator 3 (SRC3) is identified as the most active KAT for CaM. We further demonstrate that SRC3 interacts with and acetylates CaM in a Ca2+ and NMDA receptor-dependent manner. We also show that pharmacological inhibition or genetic downregulation of SRC3 impairs CaM acetylation, synaptic plasticity, and contextual fear learning in mice. Moreover, the effects of SRC3 inhibition on synaptic plasticity and fear learning could be rescued by 3KQ-CaM, a mutant form of CaM, which mimics acetylation. Together, these observations demonstrate that SRC3 acetylates CaM and regulates synaptic plasticity and learning in mice.
Collapse
|
10
|
Zhang HL, Zhao B, Yang P, Du YQ, Han W, Xu J, Yin DM. Steroid Receptor Coactivator 3 Regulates Synaptic Plasticity and Hippocampus-dependent Memory. Neurosci Bull 2021; 37:1645-1657. [PMID: 34228315 PMCID: PMC8643392 DOI: 10.1007/s12264-021-00741-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/27/2021] [Indexed: 11/28/2022] Open
Abstract
Steroid hormones play important roles in brain development and function. The signaling of steroid hormones depends on the interaction between steroid receptors and their coactivators. Although the function of steroid receptor coactivators has been extensively studied in other tissues, their functions in the central nervous system are less well investigated. In this study, we addressed the function of steroid receptor coactivator 3 (SRC3) - a member of the p160 SRC protein family that is expressed predominantly in the hippocampus. While hippocampal development was not altered in Src3+/- mice, hippocampus-dependent functions such as short-term memory and spatial memory were impaired. We further demonstrated that the deficient learning and memory in Src3+/- mice was strongly associated with the impairment of long-term potentiation (LTP) at Schaffer Collateral-CA1 synapses. Mechanistic studies indicated that Src3+/- mutation altered the composition of N-methyl-D-aspartate receptor subunits in the postsynaptic densities of hippocampal neurons. Finally, we showed that SRC3 regulated synaptic plasticity and learning mainly dependent on its lysine acetyltransferase activity. Taken together, these results reveal previously unknown functions of SRC3 in the hippocampus and thus may provide insight into how steroid hormones regulate brain function.
Collapse
Affiliation(s)
- Hai-Long Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Bing Zhao
- MOE Frontiers Center for Brain Science, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Pin Yang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Yin-Quan Du
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Wei Han
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dong-Min Yin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
11
|
Mullany LK, Lonard DM, O’Malley BW. Wound Healing-related Functions of the p160 Steroid Receptor Coactivator Family. Endocrinology 2021; 162:6042238. [PMID: 33340403 PMCID: PMC7814297 DOI: 10.1210/endocr/bqaa232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Indexed: 12/24/2022]
Abstract
Multicellular organisms have evolved sophisticated mechanisms to recover and maintain original tissue functions following injury. Injury responses require a robust transcriptomic response associated with cellular reprogramming involving complex gene expression programs critical for effective tissue repair following injury. Steroid receptor coactivators (SRCs) are master transcriptional regulators of cell-cell signaling that is integral for embryogenesis, reproduction, normal physiological function, and tissue repair following injury. Effective therapeutic approaches for facilitating improved tissue regeneration and repair will likely involve temporal and combinatorial manipulation of cell-intrinsic and cell-extrinsic factors. Pleiotropic actions of SRCs that are critical for wound healing range from immune regulation and angiogenesis to maintenance of metabolic regulation in diverse organ systems. Recent evidence derived from studies of model organisms during different developmental stages indicates the importance of the interplay of immune cells and stromal cells to wound healing. With SRCs being the master regulators of cell-cell signaling integral to physiologic changes necessary for wound repair, it is becoming clear that therapeutic targeting of SRCs provides a unique opportunity for drug development in wound healing. This review will provide an overview of wound healing-related functions of SRCs with a special focus on cellular and molecular interactions important for limiting tissue damage after injury. Finally, we review recent findings showing stimulation of SRCs following cardiac injury with the SRC small molecule stimulator MCB-613 can promote cardiac protection and inhibit pathologic remodeling after myocardial infarction.
Collapse
Affiliation(s)
- Lisa K Mullany
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Bert W O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Correspondence: Bert W. O’Malley, MD, Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston TX 77030, USA.
| |
Collapse
|
12
|
Salazar-Silva R, Dantas VLG, Alves LU, Batissoco AC, Oiticica J, Lawrence EA, Kawafi A, Yang Y, Nicastro FS, Novaes BC, Hammond C, Kague E, Mingroni-Netto RC. NCOA3 identified as a new candidate to explain autosomal dominant progressive hearing loss. Hum Mol Genet 2021; 29:3691-3705. [PMID: 33326993 PMCID: PMC7823111 DOI: 10.1093/hmg/ddaa240] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/21/2020] [Accepted: 10/15/2020] [Indexed: 12/27/2022] Open
Abstract
Hearing loss is a frequent sensory impairment in humans and genetic factors account for an elevated fraction of the cases. We have investigated a large family of five generations, with 15 reported individuals presenting non-syndromic, sensorineural, bilateral and progressive hearing loss, segregating as an autosomal dominant condition. Linkage analysis, using SNP-array and selected microsatellites, identified a region of near 13 cM in chromosome 20 as the best candidate to harbour the causative mutation. After exome sequencing and filtering of variants, only one predicted deleterious variant in the NCOA3 gene (NM_181659, c.2810C > G; p.Ser937Cys) fit in with our linkage data. RT-PCR, immunostaining and in situ hybridization showed expression of ncoa3 in the inner ear of mice and zebrafish. We generated a stable homozygous zebrafish mutant line using the CRISPR/Cas9 system. ncoa3-/- did not display any major morphological abnormalities in the ear, however, anterior macular hair cells showed altered orientation. Surprisingly, chondrocytes forming the ear cartilage showed abnormal behaviour in ncoa3-/-, detaching from their location, invading the ear canal and blocking the cristae. Adult mutants displayed accumulation of denser material wrapping the otoliths of ncoa3-/- and increased bone mineral density. Altered zebrafish swimming behaviour corroborates a potential role of ncoa3 in hearing loss. In conclusion, we identified a potential candidate gene to explain hereditary hearing loss, and our functional analyses suggest subtle and abnormal skeletal behaviour as mechanisms involved in the pathogenesis of progressive sensory function impairment.
Collapse
Affiliation(s)
- R Salazar-Silva
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, 05508-090, São Paulo, Brazil
| | - Vitor Lima Goes Dantas
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, 05508-090, São Paulo, Brazil
| | - Leandro Ucela Alves
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, 05508-090, São Paulo, Brazil
| | - Ana Carla Batissoco
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, 05508-090, São Paulo, Brazil
- Laboratório de Otorrinolaringologia/LIM32 –Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo , 01246-903, São Paulo, Brazil
| | - Jeanne Oiticica
- Laboratório de Otorrinolaringologia/LIM32 –Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo , 01246-903, São Paulo, Brazil
| | - Elizabeth A Lawrence
- School of Pharmacology, Physiology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Abdelwahab Kawafi
- School of Pharmacology, Physiology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Yushi Yang
- School of Physics, University of Bristol, Bristol, BS8 1TL, United Kingdom
- Centre for Nanoscience and Quantum Information, University of Bristol, Bristol, BS8 1FD, United Kingdom
- Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol, BS8 1FD, United Kingdom
| | - Fernanda Stávale Nicastro
- Divisão de Educação e Reabilitação dos Distúrbios da Comunicação da Pontifícia Universidade Católica de São Paulo, 04022-040, São Paulo, Brazil
| | - Beatriz Caiuby Novaes
- Divisão de Educação e Reabilitação dos Distúrbios da Comunicação da Pontifícia Universidade Católica de São Paulo, 04022-040, São Paulo, Brazil
| | - Chrissy Hammond
- School of Pharmacology, Physiology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Erika Kague
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, 05508-090, São Paulo, Brazil
- School of Pharmacology, Physiology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - R C Mingroni-Netto
- Centro de Pesquisas sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, 05508-090, São Paulo, Brazil
| |
Collapse
|
13
|
Hussein D, Dallol A, Quintas R, Schulten HJ, Alomari M, Baeesa S, Bangash M, Alghamdi F, Khan I, ElAssouli MZM, Saka M, Carracedo A, Chaudhary A, Abuzenadah A. Overlapping variants in the blood, tissues and cell lines for patients with intracranial meningiomas are predominant in stem cell-related genes. Heliyon 2020; 6:e05632. [PMID: 33305042 PMCID: PMC7710648 DOI: 10.1016/j.heliyon.2020.e05632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/19/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Bulk tissue genomic analysis of meningiomas identified common somatic mutations, however, it often excluded blood-related variants. In contrast, genomic characterisation of primary cell lines that can provide critical information regarding growth and proliferation, have been rare. In our work, we identified the variants that are present in the blood, tissues and corresponding cell lines that are likely to be predictive, tumorigenic and progressive. METHOD Whole-exome sequencing was used to identify variants and distinguish related pathways that exist in 42 blood, tissues and corresponding cell lines (BTCs) samples for patients with intracranial meningiomas. Conventional sequencing was used for the confirmation of variants. Integrative analysis of the gene expression for the corresponding samples was utilised for further interpretations. RESULTS In total, 926 BTC variants were detected, implicating 845 genes. A pathway analysis of all BTC genes with damaging variants indicated the 'cell morphogenesis involved in differentiation' stem cell-related pathway to be the most frequently affected pathway. Concordantly, five stem cell-related genes, GPRIN2, ALDH3B2, ASPN, THSD7A and SIGLEC6, showed BTC variants in at least five of the patients. Variants that were heterozygous in the blood and homozygous in the tissues or the corresponding cell lines were rare (average: 1.3 ± 0.3%), and included variants in the RUNX2 and CCDC114 genes. An analysis comparing the variants detected only in tumours with aggressive features indicated a total of 240 BTC genes, implicating the 'homophilic cell adhesion via plasma membrane adhesion molecules' pathway, and identifying the stem cell-related transcription coactivator NCOA3/AIB1/SRC3 as the most frequent BTC gene. Further analysis of the possible impact of the poly-Q mutation present in the NCOA3 gene indicated associated deregulation of 15 genes, including the up-regulation of the stem cell related SEMA3D gene and the angiogenesis related VEGFA gene. CONCLUSION Stem cell-related pathways and genes showed high prevalence in the BTC variants, and novel variants in stem cell-related genes were identified for meningioma. These variants can potentially be used as predictive, tumorigenic and progressive biomarkers for meningioma.
Collapse
Affiliation(s)
- Deema Hussein
- Neurooncology Translational Group, King Fahd Medical Research Center, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia
| | - Ashraf Dallol
- Centre of Innovation for Personalized Medicine, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rita Quintas
- Galician Foundation of Genomic Medicine-SERGAS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Hans-Juergen Schulten
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mona Alomari
- Neurooncology Translational Group, King Fahd Medical Research Center, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia
| | - Saleh Baeesa
- Division of Neurosurgery, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammed Bangash
- Division of Neurosurgery, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fahad Alghamdi
- Pathology Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ishaq Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25100, Pakistan
| | - M-Zaki Mustafa ElAssouli
- Neurooncology Translational Group, King Fahd Medical Research Center, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia
| | - Mohamad Saka
- Neurooncology Translational Group, King Fahd Medical Research Center, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia
| | - Angel Carracedo
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Galician Foundation of Genomic Medicine-SERGAS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Adeel Chaudhary
- Neurooncology Translational Group, King Fahd Medical Research Center, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia
- Centre of Innovation for Personalized Medicine, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Adel Abuzenadah
- Neurooncology Translational Group, King Fahd Medical Research Center, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia
- Centre of Innovation for Personalized Medicine, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
14
|
Taei A, Kiani T, Taghizadeh Z, Moradi S, Samadian A, Mollamohammadi S, Sharifi‐Zarchi A, Guenther S, Akhlaghpour A, Asgari Abibeiglou B, Najar‐Asl M, Karamzadeh R, Khalooghi K, Braun T, Hassani S, Baharvand H. Temporal activation of LRH-1 and RAR-γ in human pluripotent stem cells induces a functional naïve-like state. EMBO Rep 2020; 21:e47533. [PMID: 33252195 PMCID: PMC7534641 DOI: 10.15252/embr.201847533] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/13/2020] [Accepted: 07/17/2020] [Indexed: 12/19/2022] Open
Abstract
Naïve pluripotency can be established in human pluripotent stem cells (hPSCs) by manipulation of transcription factors, signaling pathways, or a combination thereof. However, differences exist in the molecular and functional properties of naïve hPSCs generated by different protocols, which include varying similarities with pre-implantation human embryos, differentiation potential, and maintenance of genomic integrity. We show here that short treatment with two chemical agonists (2a) of nuclear receptors, liver receptor homologue-1 (LRH-1) and retinoic acid receptor gamma (RAR-γ), along with 2i/LIF (2a2iL) induces naïve-like pluripotency in human cells during reprogramming of fibroblasts, conversion of pre-established hPSCs, and generation of new cell lines from blastocysts. 2a2iL-hPSCs match several defined criteria of naïve-like pluripotency and contribute to human-mouse interspecies chimeras. Activation of TGF-β signaling is instrumental for acquisition of naïve-like pluripotency by the 2a2iL induction procedure, and transient activation of TGF-β signaling substitutes for 2a to generate naïve-like hPSCs. We reason that 2a2iL-hPSCs are an easily attainable system to evaluate properties of naïve-like hPSCs and for various applications.
Collapse
Affiliation(s)
- Adeleh Taei
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
- Department of Developmental BiologyUniversity of Science and CultureTehranIran
| | - Tahereh Kiani
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Zeinab Taghizadeh
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Sharif Moradi
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Azam Samadian
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Sepideh Mollamohammadi
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Ali Sharifi‐Zarchi
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
- Computer Engineering DepartmentSharif University of TechnologyTehranIran
| | - Stefan Guenther
- Department of Cardiac Development and RemodelingMax‐Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Azimeh Akhlaghpour
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Behrouz Asgari Abibeiglou
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Mostafa Najar‐Asl
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Razieh Karamzadeh
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Keynoosh Khalooghi
- Department of Cardiac Development and RemodelingMax‐Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Thomas Braun
- Department of Cardiac Development and RemodelingMax‐Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Seyedeh‐Nafiseh Hassani
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
- Department of Developmental BiologyUniversity of Science and CultureTehranIran
| |
Collapse
|
15
|
El-Gammal Z, AlOkda A, Ali SS, Reda A, Magdeldin S, Mansour R, El-Badri N. Human-stimulated oocyte extract induces genetic and mitochondrial reprogramming of mesenchymal stromal cells. PLoS One 2020; 15:e0232759. [PMID: 32453737 PMCID: PMC7250418 DOI: 10.1371/journal.pone.0232759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 04/21/2020] [Indexed: 11/25/2022] Open
Abstract
Summary Reprogramming autologous adult cells to pluripotent cells allows for relatively safe cell replacement therapy. This can be achieved by nuclear transfer, cell fusion, or induced pluripotent stem cell technology However, the epigenetic memory of the cell is considered as a great challenge facing the complete reprograming of cells by these methods. Introducing oocyte-specific factors into differentiated cells may present a promising approach by mimicking cellular reprogramming during fertilization. Methods Human bone marrow mesenchymal stromal cells (hBM-MSCs) were cultured with different concentrations of human metaphase II (M II) oocyte extract (0.1, 1, 5, 10, 30 ng/μl). Reprogramming was assessed at various exposure times (1, 4, 7 days). Cells were tested for their proliferation rate, morphological changes, expression of pluripotency markers, expression of mesenchymal to epithelial transition markers, and mitochondrial rejuvenation. (mitochondrial localization, morphological changes, bioenergetics, transmembrane potential, and levels of reactive oxygen species, ROS). Results Treatment of human BM-MSCs with 10 ng/μl oocyte extract resulted in increased cell proliferation, which was associated with the upregulation of the pluripotency genes OCT-4, NANOG, and SOX-2 and a concomitant downregulation of mesenchymal-specific genes. MSCs exhibited small, immature round mitochondria with few swollen cristae localized proximal to the cell nucleus. This was accompanied by morphological cell changes, a metabolic shift towards oxidative phosphorylation, a high mitochondrial membrane potential, and increased ROS production. Conclusion These data show that treatment with 10 ng/μl human MII-phase oocyte extract induced genetic and mitochondrial reprogramming of human BM-MSCs to a more embryonic phenotype.
Collapse
Affiliation(s)
- Zaynab El-Gammal
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Cairo, Egypt
| | - Abdelrahman AlOkda
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Cairo, Egypt
| | - Sameh S. Ali
- Center for Aging and Associated Diseases, Helmy Institute of Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Children's Cancer Hospital, Cairo, Egypt
| | - Asmaa Reda
- Center for Aging and Associated Diseases, Helmy Institute of Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Molecular and Cellular Biology Department, Faculty of Sciences, Benha University, Benha, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Research Program, Basic Research Department, Children’s Cancer Hospital, Kappelkinger, Egypt
| | | | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Cairo, Egypt
- * E-mail:
| |
Collapse
|
16
|
Dynamic CpG methylation delineates subregions within super-enhancers selectively decommissioned at the exit from naive pluripotency. Nat Commun 2020; 11:1112. [PMID: 32111830 PMCID: PMC7048827 DOI: 10.1038/s41467-020-14916-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 02/08/2020] [Indexed: 12/29/2022] Open
Abstract
Clusters of enhancers, referred as to super-enhancers (SEs), control the expression of cell identity genes. The organisation of these clusters, and how they are remodelled upon developmental transitions remain poorly understood. Here, we report the existence of two types of enhancer units within SEs typified by distinctive CpG methylation dynamics in embryonic stem cells (ESCs). We find that these units are either prone for decommissioning or remain constitutively active in epiblast stem cells (EpiSCs), as further established in the peri-implantation epiblast in vivo. Mechanistically, we show a pivotal role for ESRRB in regulating the activity of ESC-specific enhancer units and propose that the developmentally regulated silencing of ESRRB triggers the selective inactivation of these units within SEs. Our study provides insights into the molecular events that follow the loss of ESRRB binding, and offers a mechanism by which the naive pluripotency transcriptional programme can be partially reset upon embryo implantation.
Collapse
|
17
|
Zheng X, Jia Y, Qiu L, Zeng X, Xu L, Wei M, Huang C, Liu C, Chen L, Han J. A potential target for liver cancer management, lysophosphatidic acid receptor 6 (LPAR6), is transcriptionally up-regulated by the NCOA3 coactivator. J Biol Chem 2020; 295:1474-1488. [PMID: 31914406 PMCID: PMC7008366 DOI: 10.1074/jbc.ra119.009899] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/25/2019] [Indexed: 02/05/2023] Open
Abstract
Lysophosphatidic acid receptor 6 (LPAR6) is a G protein-coupled receptor that plays critical roles in cellular morphology and hair growth. Although LPAR6 overexpression is also critical for cancer cell proliferation, its role in liver cancer tumorigenesis and the underlying mechanism are poorly understood. Here, using liver cancer and matched paracancerous tissues, as well as functional assays including cell proliferation, quantitative real-time PCR, RNA-Seq, and ChIP assays, we report that LPAR6 expression is controlled by a mechanism whereby hepatocyte growth factor (HGF) suppresses liver cancer growth. We show that high LPAR6 expression promotes cell proliferation in liver cancer. More importantly, we find that LPAR6 is transcriptionally down-regulated by HGF treatment and that its transcriptional suppression depends on nuclear receptor coactivator 3 (NCOA3). We note that enrichment of NCOA3, which has histone acetyltransferase activity, is associated with histone 3 Lys-27 acetylation (H3K27ac) at the LPAR6 locus in response to HGF treatment, indicating that NCOA3 transcriptionally regulates LPAR6 through the HGF signaling cascade. Moreover, depletion of either LPAR6 or NCOA3 significantly inhibited tumor cell growth in vitro and in vivo (in mouse tumor xenograft assays), similar to the effect of the HGF treatment. Collectively, our findings indicate an epigenetic link between LPAR6 and HGF signaling in liver cancer cells, and suggest that LPAR6 can serve as a biomarker and new strategy for therapeutic interventions for managing liver cancer.
Collapse
Affiliation(s)
- Xuan Zheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China; Department of Abdominal Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yinghui Jia
- Department of Abdominal Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lei Qiu
- Department of Abdominal Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xinyi Zeng
- Department of Abdominal Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Liangliang Xu
- Department of liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingtian Wei
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Canhua Huang
- Department of Abdominal Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Cong Liu
- Department of Paediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China.
| | - Junhong Han
- Department of Abdominal Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
18
|
CARM1 (PRMT4) Acts as a Transcriptional Coactivator during Retinoic Acid-Induced Embryonic Stem Cell Differentiation. J Mol Biol 2018; 430:4168-4182. [PMID: 30153436 PMCID: PMC6186513 DOI: 10.1016/j.jmb.2018.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/25/2018] [Accepted: 08/12/2018] [Indexed: 02/07/2023]
Abstract
Activation of the retinoic acid (RA) signaling pathway is important for controlling embryonic stem cell differentiation and development. Modulation of this pathway occurs through the recruitment of different epigenetic regulators at the retinoic acid receptors (RARs) located at RA-responsive elements and/or RA-responsive regions of RA-regulated genes. Coactivator-associated arginine methyltransferase 1 (CARM1, PRMT4) is a protein arginine methyltransferase that also functions as a transcriptional coactivator. Previous studies highlight CARM1's importance in the differentiation of different cell types. We address CARM1 function during RA-induced differentiation of murine embryonic stem cells (mESCs) using shRNA lentiviral transduction and CRISPR/Cas9 technology to deplete CARM1 in mESCs. We identify CARM1 as a novel transcriptional coactivator required for the RA-associated decrease in Rex1 (Zfp42) and for the RA induction of a subset of RA-regulated genes, including CRABP2 and NR2F1 (Coup-TF1). Furthermore, CARM1 is required for mESCs to differentiate into extraembryonic endoderm in response to RA. We next characterize the epigenetic mechanisms that contribute to RA-induced transcriptional activation of CRABP2 and NR2F1 in mESCs and show for the first time that CARM1 is required for this activation. Collectively, our data demonstrate that CARM1 is required for transcriptional activation of a subset of RA target genes, and we uncover changes in the recruitment of Suz12 and the epigenetic H3K27me3 and H3K27ac marks at gene regulatory regions for CRABP2 and NR2F1 during RA-induced differentiation.
Collapse
|
19
|
SRC-3 is involved in maintaining hematopoietic stem cell quiescence by regulation of mitochondrial metabolism in mice. Blood 2018; 132:911-923. [PMID: 29959189 DOI: 10.1182/blood-2018-02-831669] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 06/25/2018] [Indexed: 12/17/2022] Open
Abstract
Quiescence maintenance is an important property of hematopoietic stem cells (HSCs), whereas the regulatory factors and underlying mechanisms involved in HSC quiescence maintenance are not fully uncovered. Here, we show that steroid receptor coactivator 3 (SRC-3) is highly expressed in HSCs, and SRC-3-deficient HSCs are less quiescent and more proliferative, resulting in increased sensitivity to chemotherapy and irradiation. Moreover, the long-term reconstituting ability of HSCs is markedly impaired in the absence of SRC-3, and SRC-3 knockout (SRC-3-/-) mice exhibit a significant disruption of hematopoietic stem and progenitor cell homeostasis. Further investigations show that SRC-3 deficiency leads to enhanced mitochondrial metabolism, accompanied by overproduction of reactive oxygen species (ROS) in HSCs. Notably, the downstream target genes of peroxisome proliferator-activated receptor-coactivators 1α (PGC-1α) involved in the regulation of mitochondrial metabolism are significantly upregulated in SRC-3-deficient HSCs. Meanwhile, a significant decrease in the expression of histone acetyltransferase GCN5 accompanied by downregulation of PGC-1α acetylation is observed in SRC-3-null HSCs. Conversely, overexpression of GCN5 can inhibit SRC-3 deficiency-induced mitochondrial metabolism enhancement and ROS overproduction, thereby evidently rescuing the impairment of HSCs in SRC-3-/- mice. Collectively, our findings demonstrate that SRC-3 plays an important role in HSC quiescence maintenance by regulating mitochondrial metabolism.
Collapse
|
20
|
El-Gammal Z, AlOkda A, El-Badri N. Role of human oocyte-enriched factors in somatic cell reprograming. Mech Ageing Dev 2018; 175:88-99. [PMID: 29890177 DOI: 10.1016/j.mad.2018.05.009] [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: 03/08/2018] [Revised: 05/23/2018] [Accepted: 05/30/2018] [Indexed: 12/20/2022]
Abstract
Cellular reprograming paves the way for creating functional patient-specific tissues to eliminate immune rejection responses by applying the same genetic profile. However, the epigenetic memory of a cell remains a challenge facing the current reprograming methods and does not allow transcription factors to bind properly. Because somatic cells can be reprogramed by transferring their nuclear contents into oocytes, introducing specific oocyte factors into differentiated cells is considered a promising approach for mimicking the reprograming process that occurs during fertilization. Mammalian metaphase II oocyte possesses a superior capacity to epigenetically reprogram somatic cell nuclei towards an embryonic stem cell-like state than the current factor-based reprograming approaches. This may be due to the presence of specific factors that are lacking in the current factor-based reprograming approaches. In this review, we focus on studies identifying human oocyte-enriched factors aiming to understand the molecular mechanisms mediating cellular reprograming. We describe the role of oocyte-enriched factors in metabolic switch, chromatin remodelling, and global epigenetic transformation. This is critical for improving the quality of resulting reprogramed cells, which is crucial for therapeutic applications.
Collapse
Affiliation(s)
- Zaynab El-Gammal
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Egypt
| | - Abdelrahman AlOkda
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Egypt.
| |
Collapse
|
21
|
Panelo LC, Machado MS, Rubio MF, Jaworski F, Alvarado CV, Paz LA, Urtreger AJ, Vazquez E, Costas MA. High RAC3 expression levels are required for induction and maintaining of cancer cell stemness. Oncotarget 2018; 9:5848-5860. [PMID: 29464039 PMCID: PMC5814179 DOI: 10.18632/oncotarget.23635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/04/2017] [Indexed: 01/10/2023] Open
Abstract
RAC3 is a transcription coactivator, usually overexpressed in several tumors and required to maintain the pluripotency in normal stem cells. In this work we studied the association between RAC3 overexpression on cancer cell stemness and the capacity of this protein to induce cancer stem properties in non tumoral cells. We performed in vitro and in vivo experiments using two strategies: by overexpressing RAC3 in the non tumoral cell line HEK293 and by silencing RAC3 in the human colorectal epithelial cell line HCT116 by transfection. Furthermore, we analysed public repository microarrays data from human colorectal tumors in different developmental stages. We found that RAC3 overexpression was mainly associated to CD133+ side-population of colon cancer cells and also to early and advanced stages of colon cancer, involving increased expression of mesenchymal and stem markers. In turn, RAC3 silencing induced diminished tumoral properties and cancer stem cells as determined by Hoechst efflux, tumorspheres and clonogenic growth, which correlated with decreased Nanog and OCT4 expression. In non tumoral cells, RAC3 overexpression induced tumoral transformation; mesenchymal phenotype and stem markers expression. Moreover, these transformed cells generated tumors in vivo. Our results demonstrate that RAC3 is required for maintaining and induction of cancer cell stemness.
Collapse
Affiliation(s)
- Laura C Panelo
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina
| | - Mileni Soares Machado
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina
| | - María F Rubio
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina.,Laboratorio de Inflamación y Cancer, IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EGA Buenos Aires, Argentina.,Argentine National Research Council (CONICET), C1425FQB Godoy Cruz (CABA), República Argentina
| | - Felipe Jaworski
- Laboratorio de Inflamación y Cancer, IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EGA Buenos Aires, Argentina
| | - Cecilia V Alvarado
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina
| | - Leonardo A Paz
- Laboratorio de Anatomía Patológica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina
| | - Alejandro J Urtreger
- Laboratorio de Anatomía Patológica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina.,Universidad de Buenos Aires, Instituto de Oncología "Angel H Roffo", Area de Investigación, C1417DTB Buenos Aires, Argentina.,Argentine National Research Council (CONICET), C1425FQB Godoy Cruz (CABA), República Argentina
| | - Elba Vazquez
- Laboratorio de Inflamación y Cancer, IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EGA Buenos Aires, Argentina.,Argentine National Research Council (CONICET), C1425FQB Godoy Cruz (CABA), República Argentina
| | - Mónica A Costas
- Laboratorio de Biología Moleculary Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, C1427ARO Buenos Aires, Argentina.,Argentine National Research Council (CONICET), C1425FQB Godoy Cruz (CABA), República Argentina
| |
Collapse
|
22
|
Festuccia N, Owens N, Navarro P. Esrrb, an estrogen-related receptor involved in early development, pluripotency, and reprogramming. FEBS Lett 2017; 592:852-877. [DOI: 10.1002/1873-3468.12826] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/11/2017] [Accepted: 08/19/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Nicola Festuccia
- Epigenetics of Stem Cells; Department of Developmental and Stem Cell Biology; Institut Pasteur; CNRS UMR3738; Paris France
| | - Nick Owens
- Epigenetics of Stem Cells; Department of Developmental and Stem Cell Biology; Institut Pasteur; CNRS UMR3738; Paris France
| | - Pablo Navarro
- Epigenetics of Stem Cells; Department of Developmental and Stem Cell Biology; Institut Pasteur; CNRS UMR3738; Paris France
| |
Collapse
|
23
|
Abstract
The connection between genetic variation and drug response has long been explored to facilitate the optimization and personalization of cancer therapy. Crucial to the identification of drug response related genetic features is the ability to separate indirect correlations from direct correlations across abundant datasets with large number of variables. Here we analyzed proteomic and pharmacogenomic data in cancer tissues and cell lines using a global statistical model connecting protein pairs, genes and anti-cancer drugs. We estimated this model using direct coupling analysis (DCA), a powerful statistical inference method that has been successfully applied to protein sequence data to extract evolutionary signals that provide insights on protein structure, folding and interactions. We used Direct Information (DI) as a metric of connectivity between proteins as well as gene-drug pairs. We were able to infer important interactions observed in cancer-related pathways from proteomic data and predict potential connectivities in cancer networks. We also identified known and potential connections for anti-cancer drugs and gene mutations using DI in pharmacogenomic data. Our findings suggest that gene-drug connections predicted with direct couplings can be used as a reliable guide to cancer therapy and expand our understanding of the effects of gene alterations on drug efficacies.
Collapse
|
24
|
Uranishi K, Akagi T, Koide H, Yokota T. Esrrb directly binds to Gata6 promoter and regulates its expression with Dax1 and Ncoa3. Biochem Biophys Res Commun 2016; 478:1720-5. [PMID: 27601327 DOI: 10.1016/j.bbrc.2016.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 09/02/2016] [Indexed: 11/27/2022]
Abstract
Estrogen-related receptor beta (Esrrb) is expressed in embryonic stem (ES) cells and is involved in self-renewal ability and pluripotency. Previously, we found that Dax1 is associated with Esrrb and represses its transcriptional activity. Further, the disruption of the Dax1-Esrrb interaction increases the expression of the extra-embryonic endoderm marker Gata6 in ES cells. Here, we investigated the influences of Esrrb and Dax1 on Gata6 expression. Esrrb overexpression in ES cells induced endogenous Gata6 mRNA and Gata6 promoter activity. In addition, the Gata6 promoter was found to contain the Esrrb recognition motifs ERRE1 and ERRE2, and the latter was the responsive element of Esrrb. Associations between ERRE2 and Esrrb were then confirmed by biotin DNA pulldown and chromatin immunoprecipitation assays. Subsequently, we showed that Esrrb activity at the Gata6 promoter was repressed by Dax1, and although Dax1 did not bind to ERRE2, it was associated with Esrrb, which directly binds to ERRE2. In addition, the transcriptional activity of Esrrb was enhanced by nuclear receptor co-activator 3 (Ncoa3), which has recently been shown to be a binding partner of Esrrb. Finally, we showed that Dax1 was associated with Ncoa3 and repressed its transcriptional activity. Taken together, the present study indicates that the Gata6 promoter is activated by Esrrb in association with Ncoa3, and Dax1 inhibited activities of Esrrb and Ncoa3, resulting maintenance of the undifferentiated status of ES cells.
Collapse
Affiliation(s)
- Kousuke Uranishi
- Department of Stem Cell Biology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan
| | - Tadayuki Akagi
- Department of Stem Cell Biology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan.
| | - Hiroshi Koide
- Department of Stem Cell Biology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan
| | - Takashi Yokota
- Department of Stem Cell Biology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan.
| |
Collapse
|
25
|
Divekar SD, Tiek DM, Fernandez A, Riggins RB. Estrogen-related receptor β (ERRβ) - renaissance receptor or receptor renaissance? NUCLEAR RECEPTOR SIGNALING 2016; 14:e002. [PMID: 27507929 PMCID: PMC4978380 DOI: 10.1621/nrs.14002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/25/2016] [Indexed: 01/11/2023]
Abstract
Estrogen-related receptors (ERRs) are founding members of the orphan nuclear receptor (ONR) subgroup of the nuclear receptor superfamily. Twenty-seven years of study have yet to identify cognate ligands for the ERRs, though they have firmly placed ERRα and ERRγ at the intersection of cellular metabolism and oncogenesis. The pace of discovery for novel functions of ERRβ, however, has until recently been somewhat slower than that of its family members. ERRβ has also been largely ignored in summaries and perspectives of the ONR literature. Here, we provide an overview of established and emerging knowledge of ERRβ in mouse, man, and other species, highlighting unique aspects of ERRβ biology that set it apart from the other two estrogen-related receptors, with a focus on the impact of alternative splicing on the structure and function of this receptor.
Collapse
Affiliation(s)
- Shailaja D Divekar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC (SDD, DMT, AF, RBR)
| | - Deanna M Tiek
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC (SDD, DMT, AF, RBR)
| | - Aileen Fernandez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC (SDD, DMT, AF, RBR)
| | - Rebecca B Riggins
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC (SDD, DMT, AF, RBR)
| |
Collapse
|
26
|
Zhang W, Ni P, Mou C, Zhang Y, Guo H, Zhao T, Loh YH, Chen L. Cops2 promotes pluripotency maintenance by Stabilizing Nanog Protein and Repressing Transcription. Sci Rep 2016; 6:26804. [PMID: 27226076 PMCID: PMC4881025 DOI: 10.1038/srep26804] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/10/2016] [Indexed: 12/02/2022] Open
Abstract
The COP9 signalosome has been implicated in pluripotency maintenance of human embryonic stem cells. Yet, the mechanism for the COP9 signalosome to regulate pluripotency remains elusive. Through knocking down individual COP9 subunits, we demonstrate that Cops2, but not the whole COP9 signalosome, is essential for pluripotency maintenance in mouse embryonic stem cells. Down-regulation of Cops2 leads to reduced expression of pluripotency genes, slower proliferation rate, G2/M cell cycle arrest, and compromised embryoid differentiation of embryonic stem cells. Cops2 also facilitates somatic cell reprogramming. We further show that Cops2 binds to Nanog protein and prevent the degradation of Nanog by proteasome. Moreover, Cops2 functions as transcriptional corepressor to facilitate pluripotency maintenance. Altogether, our data reveal the essential role and novel mechanisms of Cops2 in pluripotency maintenance.
Collapse
Affiliation(s)
- Weiyu Zhang
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Protein Sciences and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Peiling Ni
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Protein Sciences and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chunlin Mou
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Protein Sciences and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yanqin Zhang
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Protein Sciences and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hongchao Guo
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Protein Sciences and College of Life Sciences, Nankai University, Tianjin 300071, China.,State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Protein Sciences and College of Life Sciences, Nankai University, Tianjin 300071, China.,Epigenetics and Cell Fates Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore 138673, Singapore
| | - Tong Zhao
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Protein Sciences and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuin-Han Loh
- Epigenetics and Cell Fates Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore 138673, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore
| | - Lingyi Chen
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Protein Sciences and College of Life Sciences, Nankai University, Tianjin 300071, China.,State Key Laboratory of Molecular Oncology, Cancer Institute/Hospital, Chinese Academy of Medical Sciences, Beijing 100021, China
| |
Collapse
|
27
|
Mantsoki A, Devailly G, Joshi A. Gene expression variability in mammalian embryonic stem cells using single cell RNA-seq data. Comput Biol Chem 2016; 63:52-61. [PMID: 26951854 PMCID: PMC5012374 DOI: 10.1016/j.compbiolchem.2016.02.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/01/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Gene expression heterogeneity contributes to development as well as disease progression. Due to technological limitations, most studies to date have focused on differences in mean expression across experimental conditions, rather than differences in gene expression variance. The advent of single cell RNA sequencing has now made it feasible to study gene expression heterogeneity and to characterise genes based on their coefficient of variation. METHODS We collected single cell gene expression profiles for 32 human and 39 mouse embryonic stem cells and studied correlation between diverse characteristics such as network connectivity and coefficient of variation (CV) across single cells. We further systematically characterised properties unique to High CV genes. RESULTS Highly expressed genes tended to have a low CV and were enriched for cell cycle genes. In contrast, High CV genes were co-expressed with other High CV genes, were enriched for bivalent (H3K4me3 and H3K27me3) marked promoters and showed enrichment for response to DNA damage and DNA repair. CONCLUSIONS Taken together, this analysis demonstrates the divergent characteristics of genes based on their CV. High CV genes tend to form co-expression clusters and they explain bivalency at least in part.
Collapse
Affiliation(s)
- Anna Mantsoki
- The Roslin institute, University of Edinburgh, Easter bush campus, Midlothian EH25 9RG, UK.
| | - Guillaume Devailly
- The Roslin institute, University of Edinburgh, Easter bush campus, Midlothian EH25 9RG, UK.
| | - Anagha Joshi
- The Roslin institute, University of Edinburgh, Easter bush campus, Midlothian EH25 9RG, UK.
| |
Collapse
|
28
|
Steinritz D, Schmidt A, Balszuweit F, Thiermann H, Simons T, Striepling E, Bölck B, Bloch W. Epigenetic modulations in early endothelial cells and DNA hypermethylation in human skin after sulfur mustard exposure. Toxicol Lett 2016; 244:95-102. [DOI: 10.1016/j.toxlet.2015.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/13/2015] [Accepted: 09/16/2015] [Indexed: 12/11/2022]
|
29
|
Fang L, Zhang J, Zhang H, Yang X, Jin X, Zhang L, Skalnik DG, Jin Y, Zhang Y, Huang X, Li J, Wong J. H3K4 Methyltransferase Set1a Is A Key Oct4 Coactivator Essential for Generation of Oct4 Positive Inner Cell Mass. Stem Cells 2016; 34:565-80. [PMID: 26785054 DOI: 10.1002/stem.2250] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 10/01/2015] [Indexed: 11/09/2022]
Abstract
Limited core transcription factors and transcriptional cofactors have been shown to govern embryonic stem cell (ESC) transcriptional circuitry and pluripotency, but the molecular interactions between the core transcription factors and cofactors remains ill defined. Here, we analyzed the protein-protein interactions between Oct4, Sox2, Klf4, and Myc (abbreviated as OSKM) and a large panel of cofactors. The data reveal both specific and common interactions between OSKM and cofactors. We found that among the SET1/MLL family H3K4 methyltransferases, Set1a specifically interacts with Oct4 and this interaction is independent of Wdr5. Set1a is recruited to and required for H3K4 methylation at the Oct4 target gene promoters and transcriptional activation of Oct4 target genes in ESCs, and consistently Set1a is required for ESC maintenance and induced pluripotent stem cell generation. Gene expression profiling and chromatin immunoprecipitation-seq analyses demonstrate the broad involvement of Set1a in Oct4 transcription circuitry and strong enrichment at TSS sites. Gene knockout study demonstrates that Set1a is not only required for mouse early embryonic development but also for the generation of Oct4-positive inner cell mass. Together our study provides valuable information on the molecular interactions between OSKM and cofactors and molecular mechanisms for the functional importance of Set1a in ESCs and early development.
Collapse
Affiliation(s)
- Lan Fang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jun Zhang
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University and National Resource Center for Mutant Mice, Nanjing, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hui Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiaoqin Yang
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Xueling Jin
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ling Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - David G Skalnik
- Biology Department, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Ying Jin
- Department of Molecular Development, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yong Zhang
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Xingxu Huang
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University and National Resource Center for Mutant Mice, Nanjing, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jiwen Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
30
|
Mou C, Zhang Y, Zhang W, Ding Y, Chen L. Lysine residues 639 and 673 of mouse Ncoa3 are ubiquitination sites for the regulation of its stability. Acta Biochim Biophys Sin (Shanghai) 2014; 46:1066-71. [PMID: 25348736 DOI: 10.1093/abbs/gmu096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Ncoa3 is a transcriptional coactivator involved in a wide range of biological processes. Regulation of Ncoa3 protein stability is important to control its activity precisely. Here, we found that deleting amino acid residues 614-740 of Ncoa3 enhances the protein expression level. Replacing two lysine residues, K639 and K673, within this region by arginine, increases the stability of the luciferase fusion protein as well as Ncoa3 protein. When these two lysine residues are mutated to arginine, the overall ubiquitination level of Ncoa3 decreases, indicating that lysine 639 and 673 are its ubiquitination sites. Taken together, we identified two ubiquitination sites at lysine 639 and 673 of Ncoa3. Ubiquitination of these two lysine residues leads to proteasomal degradation of Ncoa3.
Collapse
Affiliation(s)
- Chunlin Mou
- State key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yanqin Zhang
- State key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weiyu Zhang
- State key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yu Ding
- State key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lingyi Chen
- State key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center for Biotherapy, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics and College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
31
|
Dasgupta S, O'Malley BW. Transcriptional coregulators: emerging roles of SRC family of coactivators in disease pathology. J Mol Endocrinol 2014; 53:R47-59. [PMID: 25024406 PMCID: PMC4152414 DOI: 10.1530/jme-14-0080] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Transcriptional coactivators have evolved as an important new class of functional proteins that participate with virtually all transcription factors and nuclear receptors (NRs) to intricately regulate gene expression in response to a wide variety of environmental cues. Recent findings have highlighted that coactivators are important for almost all biological functions, and consequently, genetic defects can lead to severe pathologies. Drug discovery efforts targeting coactivators may prove valuable for treatment of a variety of diseases.
Collapse
Affiliation(s)
- Subhamoy Dasgupta
- Department of Molecular and Cellular BiologyBaylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular BiologyBaylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| |
Collapse
|
32
|
Du J, Wu Y, Ai Z, Shi X, Chen L, Guo Z. Mechanism of SB431542 in inhibiting mouse embryonic stem cell differentiation. Cell Signal 2014; 26:2107-16. [PMID: 24949833 DOI: 10.1016/j.cellsig.2014.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/09/2014] [Accepted: 06/09/2014] [Indexed: 12/22/2022]
Abstract
SB431542 (SB) is an established small molecular inhibitor that specifically binds to the ATP binding domains of the activin receptor-like kinase receptors, ALK5, ALK4 and ALK7, and thus specifically inhibits Smad2/3 activation and blocks TGF-β signal transduction. SB maintains the undifferentiated state of mouse embryonic stem cells. However, the way of SB in maintaining the undifferentiated state of mouse embryonic stem cells remains unclear. Considering that SB could not maintain embryonic stem cells pluripotency when leukemia inhibitory factor was withdrawn, we sought to identify the mechanism of SB on pluripotent maintenance. Transcripts regulated by SB, including message RNAs and small non-coding RNAs were examined through microarray and deep-sequence experiments. After examination, Western blot analysis, and quantitative real-time PCR verification, we found that SB regulated the transcript expressions related to self-renewal and differentiation. SB mainly functioned by inhibiting differentiation. The key pluripotent factors expression were not significantly affected by SB, and intrinsic differentiation-related transcripts including fibroblast growth factor family members, were significantly down-regulated by SB. Moreover, SB could partially inhibit the retinoic acid response to neuronal differentiation of mouse embryonic stem cells.
Collapse
Affiliation(s)
- Juan Du
- College of Life Sciences, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Yongyan Wu
- College of Veterinary Medicine, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Zhiying Ai
- College of Life Sciences, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Xiaoyan Shi
- College of Life Sciences, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Linlin Chen
- College of Life Sciences, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Zekun Guo
- College of Veterinary Medicine, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China.
| |
Collapse
|
33
|
Saunders A, Faiola F, Wang J. Concise review: pursuing self-renewal and pluripotency with the stem cell factor Nanog. Stem Cells 2014; 31:1227-36. [PMID: 23653415 DOI: 10.1002/stem.1384] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 03/04/2013] [Indexed: 01/06/2023]
Abstract
Pluripotent embryonic stem cells and induced pluripotent stem cells hold great promise for future use in tissue replacement therapies due to their ability to self-renew indefinitely and to differentiate into all adult cell types. Harnessing this therapeutic potential efficiently requires a much deeper understanding of the molecular processes at work within the pluripotency network. The transcription factors Nanog, Oct4, and Sox2 reside at the core of this network, where they interact and regulate their own expression as well as that of numerous other pluripotency factors. Of these core factors, Nanog is critical for blocking the differentiation of pluripotent cells, and more importantly, for establishing the pluripotent ground state during somatic cell reprogramming. Both mouse and human Nanog are able to form dimers in vivo, allowing them to preferentially interact with certain factors and perform unique functions. Recent studies have identified an evolutionary functional conservation among vertebrate Nanog orthologs from chick, zebrafish, and the axolotl salamander, adding an additional layer of complexity to Nanog function. Here, we present a detailed overview of published work focusing on Nanog structure, function, dimerization, and regulation at the genetic and post-translational levels with regard to the establishment and maintenance of pluripotency. The full spectrum of Nanog function in pluripotent stem cells and in cancer is only beginning to be revealed. We therefore use this evidence to advocate for more comprehensive analysis of Nanog in the context of disease, development, and regeneration.
Collapse
Affiliation(s)
- Arven Saunders
- Department of Developmental and Regenerative Biology, Black Family Stem Cell Institute, New York, NY, USA
| | | | | |
Collapse
|
34
|
Feltes BC, Bonatto D. Combining small molecules for cell reprogramming through an interatomic analysis. MOLECULAR BIOSYSTEMS 2014; 9:2741-63. [PMID: 24056910 DOI: 10.1039/c3mb70159j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The knowledge available about the application and generation of induced pluripotent stem cells (iPSC) has grown since their discovery, and new techniques to enhance the reprogramming process have been described. Among the new approaches to induce iPSC that have gained great attention is the use of small molecules for reprogramming. The application of small molecules, unlike genetic manipulation, provides for control of the reprogramming process through the shifting of concentrations and the combination of different molecules. However, different researchers have reported the use of "reprogramming cocktails" with variable results and drug combinations. Thus, the proper combination of small molecules for successful and enhanced reprogramming is a matter for discussion. However, testing all potential drug combinations in different cell lineages is very costly and time-consuming. Therefore, in this article, we discuss the use of already employed molecules for iPSC generation, followed by the application of systems chemo-biology tools to create different data sets of protein-protein (PPI) and chemical-protein (CPI) interaction networks based on the knowledge of already used and new reprogramming cocktail combinations. We further analyzed the biological processes associated with PPI-CPI networks and provided new potential protein targets to be inhibited or expressed for stem cell reprogramming. In addition, we applied a new interference analysis to prospective targets that could negatively affect the classical pluripotency-associated factors (SOX2, NANOG, KLF4 and OCT4) and thus potentially improve reprogramming protocols.
Collapse
Affiliation(s)
- Bruno César Feltes
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves 9500 - Prédio 43421 - Sala 219, Porto Alegre, Caixa Postal 15005, RS - Brazil.
| | | |
Collapse
|
35
|
Chitilian J, Thillainadesan G, Manias J, Chang W, Walker E, Isovic M, Stanford W, Torchia J. Critical Components of the Pluripotency Network Are Targets for the p300/CBP Interacting Protein (p/CIP) in Embryonic Stem Cells. Stem Cells 2014; 32:204-15. [DOI: 10.1002/stem.1564] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 08/14/2013] [Accepted: 08/23/2013] [Indexed: 01/21/2023]
Affiliation(s)
- J.M. Chitilian
- Department of Oncology; The London Regional Cancer Program and the Lawson Health Research Institute; London Ontario Canada
- Department of Biochemistry; The University of Western Ontario; London Ontario Canada
| | - G. Thillainadesan
- Department of Oncology; The London Regional Cancer Program and the Lawson Health Research Institute; London Ontario Canada
- Department of Biochemistry; The University of Western Ontario; London Ontario Canada
| | - J.L. Manias
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada
- Department of Cellular and Molecular Medicine; Faculty of Medicine; University of Ottawa; Ottawa Ontario Canada
| | - W.Y. Chang
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada
| | - E. Walker
- Centre for the Commercialization of Regenerative Medicine; Toronto Ontario Canada
| | - M. Isovic
- Department of Oncology; The London Regional Cancer Program and the Lawson Health Research Institute; London Ontario Canada
| | - W.L. Stanford
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada
- Department of Cellular and Molecular Medicine; Faculty of Medicine; University of Ottawa; Ottawa Ontario Canada
| | - J. Torchia
- Department of Oncology; The London Regional Cancer Program and the Lawson Health Research Institute; London Ontario Canada
- Department of Biochemistry; The University of Western Ontario; London Ontario Canada
| |
Collapse
|
36
|
Lee KC, Wong WK, Feng B. Decoding the Pluripotency Network: The Emergence of New Transcription Factors. Biomedicines 2013; 1:49-78. [PMID: 28548056 PMCID: PMC5423462 DOI: 10.3390/biomedicines1010049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/10/2013] [Accepted: 12/11/2013] [Indexed: 12/25/2022] Open
Abstract
Since the successful isolation of mouse and human embryonic stem cells (ESCs) in the past decades, massive investigations have been conducted to dissect the pluripotency network that governs the ability of these cells to differentiate into all cell types. Beside the core Oct4-Sox2-Nanog circuitry, accumulating regulators, including transcription factors, epigenetic modifiers, microRNA and signaling molecules have also been found to play important roles in preserving pluripotency. Among the various regulations that orchestrate the cellular pluripotency program, transcriptional regulation is situated in the central position and appears to be dominant over other regulatory controls. In this review, we would like to summarize the recent advancements in the accumulating findings of new transcription factors that play a critical role in controlling both pluripotency network and ESC identity.
Collapse
Affiliation(s)
- Kai Chuen Lee
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Room 105A, 1/F, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Shatin, N.T., Hong Kong, China.
| | - Wing Ki Wong
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Room 105A, 1/F, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Shatin, N.T., Hong Kong, China.
| | - Bo Feng
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Room 105A, 1/F, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Shatin, N.T., Hong Kong, China.
- SBS Core Laboratory, Shenzhen Research Institute, the Chinese University of Hong Kong, 4/F CUHK Shenzhen Research Institute Building, No.10, 2nd Yuexing Road, Nanshan District, Shenzhen 518057, China.
| |
Collapse
|
37
|
SILAC Proteomics of Planarians Identifies Ncoa5 as a Conserved Component of Pluripotent Stem Cells. Cell Rep 2013; 5:1142-55. [DOI: 10.1016/j.celrep.2013.10.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 08/09/2013] [Accepted: 10/21/2013] [Indexed: 12/19/2022] Open
|
38
|
Dasgupta S, Lonard DM, O'Malley BW. Nuclear receptor coactivators: master regulators of human health and disease. Annu Rev Med 2013; 65:279-92. [PMID: 24111892 DOI: 10.1146/annurev-med-051812-145316] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Transcriptional coregulators (coactivators and corepressors) have emerged as the principal modulators of the functions of nuclear receptors and other transcription factors. During the decade since the discovery of steroid receptor coactivator-1 (SRC-1), the first authentic coregulator, more than 400 coregulators have been identified and characterized, and deciphering their function has contributed significantly to our understanding of their role in human physiology. Deregulated expression of coregulators has been implicated in diverse disease states and related pathologies. The advancement of molecular technologies has enabled us to better characterize the molecular associations of the SRC family of coactivators with other protein complexes in the context of gene regulation. These continuing discoveries not only expand our knowledge of the roles of coactivators in various human diseases but allow us to discover novel coactivator-targeting strategies for therapeutic intervention in these diseases.
Collapse
Affiliation(s)
- Subhamoy Dasgupta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030;
| | | | | |
Collapse
|
39
|
Gudas LJ. Retinoids induce stem cell differentiation via epigenetic changes. Semin Cell Dev Biol 2013; 24:701-5. [PMID: 23973942 DOI: 10.1016/j.semcdb.2013.08.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 07/24/2013] [Accepted: 08/10/2013] [Indexed: 12/20/2022]
Abstract
Vitamin A (all-trans retinol) and its active metabolites, collectively called retinoids, exert potent effects on stem cell differentiation and thus, the formation of the entire organism, in part via the modulation of the epigenome. All-trans retinoic acid (RA), through binding to the retinoic acid receptors (RARs), alters interactions of the RARs with various protein components of the transcription complex at numerous genes in stem cells, and some of these protein components of the transcription complex then either place or remove epigenetic marks on histones or on DNA, altering chromatin structure and leading to an exit from the self-renewing, pluripotent stem cell state. Different epigenetic mechanisms, i.e. first, primarily H3K27me3 marks and then DNA methylation, may be employed by embryonic stem cells and other stem cells for control of early vs. late stages of cell differentiation. Creating these stable epigenetic changes requires the actions of many molecules, including tet1, polycomb protein complexes (PRCs), miRNAs, DNA methyltransferases (DNMTs), and telomerase reverse transcriptase. A more complete understanding of retinoid-dependent stem cell differentiation should reward us with new insights into the failure to maintain a differentiated state that is an essential part of neoplastic cell transformation and cancer.
Collapse
Affiliation(s)
- Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA; Department of Medicine, Weill Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
40
|
Gonzales KAU, Ng HH. Driving pluripotency and reprogramming: nuclear receptors at the helm. Semin Cell Dev Biol 2013; 24:670-8. [PMID: 23916717 DOI: 10.1016/j.semcdb.2013.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 07/16/2013] [Accepted: 07/21/2013] [Indexed: 01/30/2023]
Abstract
The identity of a cell is determined by the concerted interplay of multiple molecular modulators such as transcription factors, chromatin modifiers and signalling mediators. Among these, the transcriptional circuitry holds great influence on the specification and maintenance of a cellular state, and its perturbation can trigger a transition to another cell state. This is particularly striking in the field of pluripotency, where tempering the expression levels of one or few transcription factors is sufficient to induce the loss or acquisition of the pluripotent state. Recently, nuclear receptors, a class of transcription factors, have emerged as major players in the molecular network governing pluripotency. In this review, we discuss the importance of nuclear receptors in embryonic stem cell self-renewal, differentiation and cellular reprogramming, highlighting recent discoveries as well as providing an outlook in stem cell and nuclear receptor research.
Collapse
Affiliation(s)
- Kevin Andrew Uy Gonzales
- Gene Regulation Laboratory, Genome Institute of Singapore, Singapore 138672, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456, Singapore.
| | | |
Collapse
|
41
|
Xu Z, Jiang J, Xu C, Wang Y, Sun L, Guo X, Liu H. MicroRNA-181 regulates CARM1 and histone arginine methylation to promote differentiation of human embryonic stem cells. PLoS One 2013; 8:e53146. [PMID: 23301034 PMCID: PMC3536801 DOI: 10.1371/journal.pone.0053146] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/23/2012] [Indexed: 12/03/2022] Open
Abstract
As a novel epigenetic mechanism, histone H3 methylation at R17 and R26, which is mainly catalyzed by coactivator-associated protein arginine methyltransferase 1 (CARM1), has been reported to modulate the transcription of key pluripotency factors and to regulate pluripotency in mouse embryos and mouse embryonic stem cells (mESCs) in previous studies. However, the role of CARM1 in human embryonic stem cells (hESCs) and the regulatory mechanism that controls CARM1 expression during ESCs differentiation are presently unknown. Here, we demonstrate that CARM1 plays an active role in the resistance to differentiation in hESCs by regulating pluripotency genes in response to BMP4. In a functional screen, we identified the miR-181 family as a regulator of CARM1 that is induced during ESC differentiation and show that endogenous miR-181c represses the expression of CARM1. Depletion of CARM1 or enforced expression of miR-181c inhibits the expression of pluripotency genes and induces differentiation independent of BMP4, whereas overexpression of CARM1 or miR-181c inhibitor elevates Nanog and impedes differentiation. Furthermore, expression of CARM1 rescue constructs inhibits the effect of miR-181c overexpression in promoting differentiation. Taken together, our findings demonstrate the importance of a miR-181c-CARM1 pathway in regulating the differentiation of hESCs.
Collapse
Affiliation(s)
- Zhenyu Xu
- Research Center of Developmental Biology, Second Military Medical University, Shanghai, China
| | - Junfeng Jiang
- Research Center of Developmental Biology, Second Military Medical University, Shanghai, China
| | - Chen Xu
- Research Center of Developmental Biology, Second Military Medical University, Shanghai, China
| | - Yue Wang
- Research Center of Developmental Biology, Second Military Medical University, Shanghai, China
- * E-mail: (HL); (YW)
| | - Lei Sun
- Research Center of Developmental Biology, Second Military Medical University, Shanghai, China
| | - Xiaocan Guo
- Research Center of Developmental Biology, Second Military Medical University, Shanghai, China
| | - Houqi Liu
- Research Center of Developmental Biology, Second Military Medical University, Shanghai, China
- * E-mail: (HL); (YW)
| |
Collapse
|
42
|
Percharde M, Azuara V. Essential roles for the nuclear receptor coactivator Ncoa3 in pluripotency. Cell Cycle 2012; 12:195-6. [PMID: 23287469 PMCID: PMC3575442 DOI: 10.4161/cc.23377] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|