1
|
Xiao J, You K, Lu D, Guan S, Wu H, Gao J, Tang Y, Yu S, Gao B. Cell-Derived Basal Membrane-Like Extracellular Matrix Promotes Endothelial Cell Expansion and Functionalization. J Biomed Mater Res A 2025; 113:e37893. [PMID: 40059713 DOI: 10.1002/jbm.a.37893] [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: 10/23/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
Engineering cellular microenvironments with biomaterials is an effective strategy for endothelial cell expansion and functionality in vascular tissue engineering. The basement membrane (BM) is a natural vascular endothelium microenvironment that plays an important role in promoting rapid expansion and function of endothelial cells. However, mimicking the crucial function of BM with an ideal biomaterial remains challenging. In this study, we developed a cell-derived decellularized extracellular matrix (c-dECM) paper to mimic the role of BM in endothelial cell expansion and function. The results showed that c-dECM paper was a stable, biocompatible, and biodegradable scaffold that significantly promoted endothelial cell expansion by modulating cell migration, adhesion, and proliferation both in vivo and in vitro. Moreover, the biomimetic c-dECM paper can profoundly promote endothelial cell function by increasing the synthesis and release of nitric oxide (NO) and prostaglandin I2 (PGI2) and upregulating the expression of anticoagulant and vascularized genes, including thrombomodulin (THBD), tissue factor pathway inhibitor (TFPI), endothelial growth factor (VEGF) and endoglin (CD105). These data indicate that the c-dECM is a potential biomaterial for constructing vascular tissue engineering scaffolds or developing in vitro models to study the functional mechanisms of endothelial cells.
Collapse
Affiliation(s)
- Jiangwei Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Kai You
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Daohuan Lu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Shuwen Guan
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Hengpeng Wu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Jing Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Yadong Tang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Shan Yu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Chemical Engineering Department, Ningbo Key Laboratory of High Performance Petroleum Resin Preparation Engineering and Technology, Ningbo, China
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
2
|
Athanasouli P, Vanhessche T, Lluis F. Divergent destinies: insights into the molecular mechanisms underlying EPI and PE fate determination. Life Sci Alliance 2025; 8:e202403091. [PMID: 39779220 PMCID: PMC11711469 DOI: 10.26508/lsa.202403091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Mammalian pre-implantation development is entirely devoted to the specification of extra-embryonic lineages, which are fundamental for embryo morphogenesis and support. The second fate decision is taken just before implantation, as defined by the epiblast (EPI) and the primitive endoderm (PE) specification. Later, EPI forms the embryo proper and PE contributes to the formation of the yolk sac. The formation of EPI and PE as molecularly and morphologically distinct lineages is the final step of a multistage process, which begins when bipotent progenitor cells diverge into separate fates. Despite advances in uncovering the molecular mechanisms underlying the differential transcriptional patterns that dictate how apparently identical cells make fate decisions and how lineage integrity is maintained, a detailed overview of these mechanisms is still lacking. In this review, we dissect the EPI and PE formation process into four stages (initiation, specification, segregation, and maintenance) and we provide a comprehensive understanding of the molecular mechanisms involved in lineage establishment in the mouse. In addition, we discuss the conservation of key processes in humans, based on the most recent findings.
Collapse
Affiliation(s)
- Paraskevi Athanasouli
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Tijs Vanhessche
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Frederic Lluis
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Chousal JN, Morey R, Srinivasan S, Lee K, Zhang W, Yeo AL, To C, Cho K, Garzo VG, Parast MM, Laurent LC, Cook-Andersen H. Molecular profiling of human blastocysts reveals primitive endoderm defects among embryos of decreased implantation potential. Cell Rep 2024; 43:113701. [PMID: 38277271 DOI: 10.1016/j.celrep.2024.113701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 12/12/2023] [Accepted: 01/05/2024] [Indexed: 01/28/2024] Open
Abstract
Human embryo implantation is remarkably inefficient, and implantation failure remains among the greatest obstacles in treating infertility. Gene expression data from human embryos have accumulated rapidly in recent years; however, identification of the subset of genes that determine successful implantation remains a challenge. We leverage clinical morphologic grading-known for decades to correlate with implantation potential-and transcriptome analyses of matched embryonic and abembryonic samples to identify factors and pathways enriched and depleted in human blastocysts of good and poor morphology. Unexpectedly, we discovered that the greatest difference was in the state of extraembryonic primitive endoderm (PrE) development, with relative deficiencies in poor morphology blastocysts. Our results suggest that implantation success is most strongly influenced by the embryonic compartment and that deficient PrE development is common among embryos with decreased implantation potential. Our study provides a valuable resource for those investigating the markers and mechanisms of human embryo implantation.
Collapse
Affiliation(s)
- Jennifer N Chousal
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert Morey
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Srimeenakshi Srinivasan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Katherine Lee
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wei Zhang
- Reproductive Partners Fertility Center - San Diego, La Jolla, CA 92037, USA
| | - Ana Lisa Yeo
- Reproductive Partners Fertility Center - San Diego, La Jolla, CA 92037, USA
| | - Cuong To
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kyucheol Cho
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - V Gabriel Garzo
- Reproductive Partners Fertility Center - San Diego, La Jolla, CA 92037, USA
| | - Mana M Parast
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Heidi Cook-Andersen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
4
|
Perera M, Brickman JM. In vitro models of human hypoblast and mouse primitive endoderm. Curr Opin Genet Dev 2023; 83:102115. [PMID: 37783145 DOI: 10.1016/j.gde.2023.102115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/28/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023]
Abstract
The primitive endoderm (PrE, also named hypoblast), a predominantly extraembryonic epithelium that arises from the inner cell mass (ICM) of the mammalian pre-implantation blastocyst, plays a fundamental role in embryonic development, giving rise to the yolk sac, establishing the anterior-posterior axis and contributing to the gut. PrE is specified from the ICM at the same time as the epiblast (Epi) that will form the embryo proper. While in vitro cell lines resembling the pluripotent Epi have been derived from a variety of conditions, only one model system currently exists for the PrE, naïve extraembryonic endoderm (nEnd). As a result, considerably more is known about the gene regulatory networks and signalling requirements of pluripotent stem cells than nEnd. In this review, we describe the ontogeny and differentiation of the PrE or hypoblast in mouse and primate and then discuss in vitro cell culture models for different extraembryonic endodermal cell types.
Collapse
Affiliation(s)
- Marta Perera
- reNEW UCPH - The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark. https://twitter.com/@MartaPrera
| | - Joshua M Brickman
- reNEW UCPH - The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| |
Collapse
|
5
|
Pham PD, Lu H, Han H, Zhou JJ, Madan A, Wang W, Murre C, Cho KWY. Transcriptional network governing extraembryonic endoderm cell fate choice. Dev Biol 2023; 502:20-37. [PMID: 37423592 PMCID: PMC10550205 DOI: 10.1016/j.ydbio.2023.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023]
Abstract
The mechanism by which transcription factor (TF) network instructs cell-type-specific transcriptional programs to drive primitive endoderm (PrE) progenitors to commit to parietal endoderm (PE) versus visceral endoderm (VE) cell fates remains poorly understood. To address the question, we analyzed the single-cell transcriptional signatures defining PrE, PE, and VE cell states during the onset of the PE-VE lineage bifurcation. By coupling with the epigenomic comparison of active enhancers unique to PE and VE cells, we identified GATA6, SOX17, and FOXA2 as central regulators for the lineage divergence. Transcriptomic analysis of cXEN cells, an in vitro model for PE cells, after the acute depletion of GATA6 or SOX17 demonstrated that these factors induce Mycn, imparting the self-renewal properties of PE cells. Concurrently, they suppress the VE gene program, including key genes like Hnf4a and Ttr, among others. We proceeded with RNA-seq analysis on cXEN cells with FOXA2 knockout, in conjunction with GATA6 or SOX17 depletion. We found FOXA2 acts as a potent suppressor of Mycn while simultaneously activating the VE gene program. The antagonistic gene regulatory activities of GATA6/SOX17 and FOXA2 in promoting alternative cell fates, and their physical co-bindings at the enhancers provide molecular insights to the plasticity of the PrE lineage. Finally, we show that the external cue, BMP signaling, promotes the VE cell fate by activation of VE TFs and repression of PE TFs including GATA6 and SOX17. These data reveal a putative core gene regulatory module that underpins PE and VE cell fate choice.
Collapse
Affiliation(s)
- Paula Duyen Pham
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Hanbin Lu
- School of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, CA, 92039, USA
| | - Han Han
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Jeff Jiajing Zhou
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Aarushi Madan
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Cornelis Murre
- School of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, CA, 92039, USA
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
6
|
Chiang IKN, Graus MS, Kirschnick N, Davidson T, Luu W, Harwood R, Jiang K, Li B, Wong YY, Moustaqil M, Lesieur E, Skoczylas R, Kouskoff V, Kazenwadel J, Arriola‐Martinez L, Sierecki E, Gambin Y, Alitalo K, Kiefer F, Harvey NL, Francois M. The blood vasculature instructs lymphatic patterning in a SOX7-dependent manner. EMBO J 2023; 42:e109032. [PMID: 36715213 PMCID: PMC9975944 DOI: 10.15252/embj.2021109032] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 01/31/2023] Open
Abstract
Despite a growing catalog of secreted factors critical for lymphatic network assembly, little is known about the mechanisms that modulate the expression level of these molecular cues in blood vascular endothelial cells (BECs). Here, we show that a BEC-specific transcription factor, SOX7, plays a crucial role in a non-cell-autonomous manner by modulating the transcription of angiocrine signals to pattern lymphatic vessels. While SOX7 is not expressed in lymphatic endothelial cells (LECs), the conditional loss of SOX7 function in mouse embryos causes a dysmorphic dermal lymphatic phenotype. We identify novel distant regulatory regions in mice and humans that contribute to directly repressing the transcription of a major lymphangiogenic growth factor (Vegfc) in a SOX7-dependent manner. Further, we show that SOX7 directly binds HEY1, a canonical repressor of the Notch pathway, suggesting that transcriptional repression may also be modulated by the recruitment of this protein partner at Vegfc genomic regulatory regions. Our work unveils a role for SOX7 in modulating downstream signaling events crucial for lymphatic patterning, at least in part via the transcriptional repression of VEGFC levels in the blood vascular endothelium.
Collapse
Affiliation(s)
- Ivy K N Chiang
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Matthew S Graus
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Nils Kirschnick
- European Institute for Molecular Imaging (EIMI)University of MünsterMünsterGermany
| | - Tara Davidson
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Winnie Luu
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Richard Harwood
- Sydney Microscopy and MicroanalysisUniversity of SydneySydneyNSWAustralia
| | - Keyi Jiang
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Bitong Li
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
| | - Yew Yan Wong
- The Genome Imaging CenterThe Centenary InstituteSydneyNSWAustralia
| | - Mehdi Moustaqil
- EMBL Australia Node in Single Molecule Science, and School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Emmanuelle Lesieur
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaQLDAustralia
| | - Renae Skoczylas
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaQLDAustralia
| | - Valerie Kouskoff
- Division of Developmental Biology & MedicineThe University of ManchesterManchesterUK
| | - Jan Kazenwadel
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSAAustralia
| | - Luis Arriola‐Martinez
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSAAustralia
| | - Emma Sierecki
- EMBL Australia Node in Single Molecule Science, and School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Yann Gambin
- EMBL Australia Node in Single Molecule Science, and School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Friedmann Kiefer
- European Institute for Molecular Imaging (EIMI)University of MünsterMünsterGermany
| | - Natasha L Harvey
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSAAustralia
| | - Mathias Francois
- The Centenary Institute, David Richmond Program for Cardio‐Vascular Research: Gene Regulation and Editing, Sydney Medical SchoolUniversity of SydneySydneyNSWAustralia
- The Genome Imaging CenterThe Centenary InstituteSydneyNSWAustralia
| |
Collapse
|
7
|
Rapid induction and long-term self-renewal of neural crest-derived ectodermal chondrogenic cells from hPSCs. NPJ Regen Med 2022; 7:69. [PMID: 36477591 PMCID: PMC9729200 DOI: 10.1038/s41536-022-00265-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Articular cartilage is highly specific and has limited capacity for regeneration if damaged. Human pluripotent stem cells (hPSCs) have the potential to generate any cell type in the body. Here, we report the dual-phase induction of ectodermal chondrogenic cells (ECCs) from hPSCs through the neural crest (NC). ECCs were able to self-renew long-term (over numerous passages) in a cocktail of growth factors and small molecules. The cells stably expressed cranial neural crest-derived mandibular condylar cartilage markers, such as MSX1, FOXC1 and FOXC2. Compared with chondroprogenitors from iPSCs via the paraxial mesoderm, ECCs had single-cell transcriptome profiles similar to condylar chondrocytes. After the removal of the cocktail sustaining self-renewal, the cells stopped proliferating and differentiated into a homogenous chondrocyte population. Remarkably, after transplantation, this cell lineage was able to form cartilage-like structures resembling mandibular condylar cartilage in vivo. This finding provides a framework to generate self-renewing cranial chondrogenic progenitors, which could be useful for developing cell-based therapy for cranial cartilage injury.
Collapse
|
8
|
Chowdhary S, Hadjantonakis AK. Journey of the mouse primitive endoderm: from specification to maturation. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210252. [PMID: 36252215 PMCID: PMC9574636 DOI: 10.1098/rstb.2021.0252] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/25/2022] [Indexed: 12/22/2022] Open
Abstract
The blastocyst is a conserved stage and distinct milestone in the development of the mammalian embryo. Blastocyst stage embryos comprise three cell lineages which arise through two sequential binary cell fate specification steps. In the first, extra-embryonic trophectoderm (TE) cells segregate from inner cell mass (ICM) cells. Subsequently, ICM cells acquire a pluripotent epiblast (Epi) or extra-embryonic primitive endoderm (PrE, also referred to as hypoblast) identity. In the mouse, nascent Epi and PrE cells emerge in a salt-and-pepper distribution in the early blastocyst and are subsequently sorted into adjacent tissue layers by the late blastocyst stage. Epi cells cluster at the interior of the ICM, while PrE cells are positioned on its surface interfacing the blastocyst cavity, where they display apicobasal polarity. As the embryo implants into the maternal uterus, cells at the periphery of the PrE epithelium, at the intersection with the TE, break away and migrate along the TE as they mature into parietal endoderm (ParE). PrE cells remaining in association with the Epi mature into visceral endoderm. In this review, we discuss our current understanding of the PrE from its specification to its maturation. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.
Collapse
Affiliation(s)
- Sayali Chowdhary
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
9
|
Filimonow K, de la Fuente R. Specification and role of extraembryonic endoderm lineages in the periimplantation mouse embryo. Theriogenology 2021; 180:189-206. [PMID: 34998083 DOI: 10.1016/j.theriogenology.2021.12.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022]
Abstract
During mammalian embryo development, the correct formation of the first extraembryonic endoderm lineages is fundamental for successful development. In the periimplantation blastocyst, the primitive endoderm (PrE) is formed, which gives rise to the parietal endoderm (PE) and visceral endoderm (VE) during further developmental stages. These PrE-derived lineages show significant differences in both their formation and roles. Whereas differentiation of the PE as a migratory lineage has been suggested to represent the first epithelial-to-mesenchymal transition (EMT) in development, organisation of the epithelial VE is of utmost importance for the correct axis definition and patterning of the embryo. Despite sharing a common origin, the striking differences between the VE and PE are indicative of their distinct roles in early development. However, there is a significant disparity in the current knowledge of each lineage, which reflects the need for a deeper understanding of their respective specification processes. In this review, we will discuss the origin and maturation of the PrE, PE, and VE during the periimplantation period using the mouse model as an example. Additionally, we consider the latest findings regarding the role of the PrE-derived lineages and early embryo morphogenesis, as obtained from the most recent in vitro models.
Collapse
Affiliation(s)
- Katarzyna Filimonow
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland.
| | - Roberto de la Fuente
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland.
| |
Collapse
|
10
|
Sun-Wada GH, Tabata H, Wada Y. Vacuolar-type proton ATPase is required for maintenance of apicobasal polarity of embryonic visceral endoderm. Sci Rep 2021; 11:19355. [PMID: 34588579 PMCID: PMC8481250 DOI: 10.1038/s41598-021-98952-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/17/2021] [Indexed: 12/29/2022] Open
Abstract
The endocytic compartments keep their interior acidic through the inward flow of protons and anions from the cytosol. Acidification is mediated by a proton pump known as vacuolar-type ATPase (V-ATPase) and transporters conferring anion conductance to the organellar membrane. In this study, we analysed the phenotype of mouse embryos lacking the V-ATPase c-subunit. The mutant embryos differentiated embryonic epithelial tissues, primitive endoderm, epiblast, and extraembryonic ectoderm; however, the organisation of these epithelia was severely affected. The apical-basal polarity in the visceral endoderm layer was not properly established in the mutant embryos, resulting in abnormal epithelial morphology. Thus, the function of V-ATPase is imperative for the establishment and/or maintenance of epithelial cell polarity, which is required for early embryogenesis.
Collapse
Affiliation(s)
- Ge-Hong Sun-Wada
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kohdo, Kyotanabe, Kyoto, 610-0395, Japan
| | - Hiroyuki Tabata
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kohdo, Kyotanabe, Kyoto, 610-0395, Japan.,Division of Biochemistry, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kami-ohno, Himeji, Hyogo, 670-8524, Japan
| | - Yoh Wada
- Division of Biological Sciences, Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka, 567-0047, Japan.
| |
Collapse
|
11
|
Hong N, Zhang E, Xie H, Jin L, Zhang Q, Lu Y, Chen AF, Yu Y, Zhou B, Chen S, Yu Y, Sun K. The transcription factor Sox7 modulates endocardiac cushion formation contributed to atrioventricular septal defect through Wnt4/Bmp2 signaling. Cell Death Dis 2021; 12:393. [PMID: 33846290 PMCID: PMC8041771 DOI: 10.1038/s41419-021-03658-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 03/22/2021] [Indexed: 02/02/2023]
Abstract
Cardiac septum malformations account for the largest proportion in congenital heart defects. The transcription factor Sox7 has critical functions in the vascular development and angiogenesis. It is unclear whether Sox7 also contributes to cardiac septation development. We identified a de novo 8p23.1 deletion with Sox7 haploinsufficiency in an atrioventricular septal defect (AVSD) patient using whole exome sequencing in 100 AVSD patients. Then, multiple Sox7 conditional loss-of-function mice models were generated to explore the role of Sox7 in atrioventricular cushion development. Sox7 deficiency mice embryos exhibited partial AVSD and impaired endothelial to mesenchymal transition (EndMT). Transcriptome analysis revealed BMP signaling pathway was significantly downregulated in Sox7 deficiency atrioventricular cushions. Mechanistically, Sox7 deficiency reduced the expressions of Bmp2 in atrioventricular canal myocardium and Wnt4 in endocardium, and Sox7 binds to Wnt4 and Bmp2 directly. Furthermore, WNT4 or BMP2 protein could partially rescue the impaired EndMT process caused by Sox7 deficiency, and inhibition of BMP2 by Noggin could attenuate the effect of WNT4 protein. In summary, our findings identify Sox7 as a novel AVSD pathogenic candidate gene, and it can regulate the EndMT involved in atrioventricular cushion morphogenesis through Wnt4-Bmp2 signaling. This study contributes new strategies to the diagnosis and treatment of congenital heart defects.
Collapse
Affiliation(s)
- Nanchao Hong
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China ,grid.8547.e0000 0001 0125 2443Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Erge Zhang
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Huilin Xie
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China ,grid.8547.e0000 0001 0125 2443Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Lihui Jin
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Qi Zhang
- grid.16821.3c0000 0004 0368 8293Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092 Shanghai, China
| | - Yanan Lu
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Alex F. Chen
- grid.16821.3c0000 0004 0368 8293Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092 Shanghai, China
| | - Yongguo Yu
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Bin Zhou
- grid.9227.e0000000119573309Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China
| | - Sun Chen
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Yu Yu
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China ,grid.16821.3c0000 0004 0368 8293Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092 Shanghai, China
| | - Kun Sun
- grid.16821.3c0000 0004 0368 8293Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| |
Collapse
|
12
|
Olbromski M, Podhorska-Okołów M, Dzięgiel P. Role of SOX Protein Groups F and H in Lung Cancer Progression. Cancers (Basel) 2020; 12:cancers12113235. [PMID: 33152990 PMCID: PMC7692225 DOI: 10.3390/cancers12113235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The expression of SOX proteins has been demonstrated in many tissues at various stages of embryogenesis, where they play the role of transcription factors. The SOX18 protein (along with SOX7 and SOX17) belongs to the SOXF group and is mainly involved in the development of the cardiovascular system, where its expression was found in the endothelium. SOX18 expression was also demonstrated in neoplastic lines of gastric, pancreatic and colon adenocarcinomas. The prognostic role of SOX30 expression has only been studied in lung adenocarcinomas, where a low expression of this factor in the stromal tumor was associated with a worse prognosis for patients. Because of the complexity of non-small-cell lung cancer (NSCLC) development, the role of the SOX proteins in this malignancy is still not fully understood. Many recently published papers show that SOX family protein members play a crucial role in the progression of NSCLC. Abstract The SOX family proteins are proved to play a crucial role in the development of the lymphatic ducts and the cardiovascular system. Moreover, an increased expression level of the SOX18 protein has been found in many malignances, such as melanoma, stomach, pancreatic breast and lung cancers. Another SOX family protein, the SOX30 transcription factor, is responsible for the development of male germ cells. Additionally, recent studies have shown its proapoptotic character in non-small cell lung cancer cells. Our preliminary studies showed a disparity in the amount of mRNA of the SOX18 gene relative to the amount of protein. This is why our attention has been focused on microRNA (miRNA) molecules, which could regulate the SOX18 gene transcript level. Recent data point to the fact that, in practically all types of cancer, hundreds of genes exhibit an abnormal methylation, covering around 5–10% of the thousands of CpG islands present in the promoter sequences, which in normal cells should not be methylated from the moment the embryo finishes its development. It has been demonstrated that in non-small-cell lung cancer (NSCLC) cases there is a large heterogeneity of the methylation process. The role of the SOX18 and SOX30 expression in non-small-cell lung cancers (NSCLCs) is not yet fully understood. However, if we take into account previous reports, these proteins may be important factors in the development and progression of these malignancies.
Collapse
Affiliation(s)
- Mateusz Olbromski
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Medical University, 50-368 Wroclaw, Poland;
- Correspondence: ; Tel.: +48-717-841-354; Fax: +48-717-840-082
| | - Marzenna Podhorska-Okołów
- Department of Ultrastructural Research, Department of Human Morphology and Embryology, Medical University, 50-368 Wroclaw, Poland;
| | - Piotr Dzięgiel
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Medical University, 50-368 Wroclaw, Poland;
- Department of Physiotherapy, University School of Physical Education, 51-612 Wroclaw, Poland
| |
Collapse
|
13
|
Yang XF, Zhou SY, Wang C, Huang W, Li N, He F, Li FR. Inhibition of LSD1 promotes the differentiation of human induced pluripotent stem cells into insulin-producing cells. Stem Cell Res Ther 2020; 11:185. [PMID: 32430053 PMCID: PMC7238565 DOI: 10.1186/s13287-020-01694-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human induced pluripotent stem cells (hiPSCs) represent a potentially unlimited source of pancreatic endocrine lineage cells. Although insulin-producing β cells derived from hiPSCs have been successfully induced, much work remains to be done to achieve mature β cells. Lysine-specific demethylase 1 (LSD1) plays an important role in the regulation of hiPSC self-renewal and differentiation. We propose a new strategy to acquire insulin-producing cells (IPCs) from hiPSCs by knocking down LSD1. METHODS Knockdown of LSD1 in hiPSCs with five shRNA. Assessment of the effects of shRNA on hiPSC proliferation, cell cycle, and apoptosis. Using knockdown hiPSCs with 31.33% LSD1 activity, we achieved a four-step differentiation into IPCs and test its differentiation efficiency, morphology, and marker genes and proteins. We implanted the IPCs into the renal subcapsular of SCID-Beige diabetic mice to evaluate the hypoglycemic effect in vivo. We tested LSD1 and HDAC1 whether they are present in the CoREST complex through IP-WB, and analyzed LSD1, CoREST, HDAC1, H3K4me2/me3, and H3K27me3 protein expression before and after knockdown of LSD1. RESULTS Differentiated hiPSCs were 38.32% ± 3.54% insulin-positive cells and released insulin/C-peptide in response to glucose stimulus in a manner comparable to adult human islets. Most of the IPCs co-expressed mature β cell-specific markers. When transplanted under the left renal capsule of SCID-Beige diabetic mice, these IPCs reversed hyperglycemia, leading to a significant increase in the definitive endoderm cells. IP-WB results showed that LSD1, HDAC1, and CoREST formed a complex in hiPSCs. Chip-PCR results showed that LSD1, HDAC1, and CoREST were enriched in the same district during the SOX17 and FOXA2 promoter region. Inhibition of LSD1 would not affect the level of CoREST but decreased the HDAC1 expressions. The H3K4me2/me3 and H3K9act level of SOX17 and FOXA2 promoter region increased after inhibited of LSD1, and promoted transcriptional activation. The H3K4me2/me3 and H3K9act level of OCT4 and SOX2 promoter region decreased with the transcriptional repressed. CONCLUSIONS LSD1 regulated histone methylation and acetylation in promoter regions of pluripotent or endodermal genes. Our results suggest a highly efficient approach to producing IPCs from hiPSCs.
Collapse
Affiliation(s)
- Xiao-Fei Yang
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China.,Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China.,Shenzhen Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Shu-Yan Zhou
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China.,Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China
| | - Ce Wang
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China.,Shenzhen Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Wei Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Ning Li
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China.,Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China
| | - Fei He
- Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China.,Shenzhen Cell Therapy Public Service Platform, Shenzhen, 518020, China
| | - Fu-Rong Li
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Ji'nan University, 1017 Dongmen North Road, Shenzhen, 518020, China. .,Guangdong Engineering Technology Research Center of Stem Cell and Cell therapy, Shenzhen, 518020, China. .,Shenzhen Cell Therapy Public Service Platform, Shenzhen, 518020, China.
| |
Collapse
|
14
|
Yang J, Hu Y, Han J, Xiao K, Liu X, Tan C, Zeng Q, Du H. Genome-wide analysis of the Chinese sturgeon sox gene family: identification, characterisation and expression profiles of different tissues. JOURNAL OF FISH BIOLOGY 2020; 96:175-184. [PMID: 31713865 DOI: 10.1111/jfb.14199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 11/11/2019] [Indexed: 06/10/2023]
Abstract
The sox family is assumed to be responsible for a number of developmental systems. Genome sequencing technology makes it possible to scan sox genes and conduct characteristic analyses of different species. In fish, full characterisation of sox genes at the genome-wide level has been reported for pufferfish Takifugu rubripes, medaka Oryzias latipes, tilapia Oreochromis niloticus and channel catfish Ictalurus punctatus. However, no systematic investigation of the sox family in sturgeons (Acipenseridae) has been reported to date. This study conducted genome-wide identification of the sox genes in the Chinese sturgeon Acipenser sinensis and profiled their tissue distribution between male and female individuals. In total, 19 sox genes were identified, including soxb1, b2, c, d, e, f and h, in the Chinese sturgeon. Genomic structure analysis indicated relatively conserved exon-intron structures in each sox group and phylogenetic analysis supported the previous classification of the sox family. Most of the sox genes showed a tissue-specific expression pattern, indicating the possible involvement of Chinese sturgeon sox genes at different developmental processes such as cardiac and gonadal development. This study provides a comprehensive resource of Chinese sturgeon sox genes and enables a better understanding of the evolution and function of the sox family.
Collapse
Affiliation(s)
- Jing Yang
- Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
| | - Yacheng Hu
- Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
| | - Jilu Han
- China Three Gorges Corporation, Yichang, China
| | - Kan Xiao
- Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
| | - Xueqing Liu
- Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
| | - Chun Tan
- Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
| | - Qingkai Zeng
- Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
| | - Hejun Du
- Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, China
| |
Collapse
|
15
|
SOX7 Target Genes and Their Contribution to Its Tumor Suppressive Function. Int J Mol Sci 2018; 19:ijms19051451. [PMID: 29757932 PMCID: PMC5983648 DOI: 10.3390/ijms19051451] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/07/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022] Open
Abstract
SOX7 is a transcription factor and acts as a tumor suppressor, but its target genes in cancers are poorly explored. We revealed SOX7-mediated gene expression profile in breast cancer cells using microarray chips and discovered multiple altered signaling pathways. When combinatorially analyzing the microarray data with a gene array dataset from 759 breast cancer patients, we identified four genes as potential targets of SOX7 and validated them by quantitative PCR and chromatin immunoprecipitation assays. Among these four genes, we determined that SOX7-activated SPRY1 and SLIT2, and SOX7-repressed TRIB3 and MTHFD2 could all differentially contribute to SOX7-mediated tumor suppression. Overall, we identified multiple cancer-related pathways mediated by SOX7 and for the first time revealed SOX7-regulated target genes in a cancer-relevant context.
Collapse
|
16
|
Rajgara RF, Lala-Tabbert N, Marchildon F, Lamarche É, MacDonald JK, Scott DA, Blais A, Skerjanc IS, Wiper-Bergeron N. SOX7 Is Required for Muscle Satellite Cell Development and Maintenance. Stem Cell Reports 2017; 9:1139-1151. [PMID: 28943254 PMCID: PMC5639291 DOI: 10.1016/j.stemcr.2017.08.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 01/26/2023] Open
Abstract
Satellite cells are skeletal-muscle-specific stem cells that are activated by injury to proliferate, differentiate, and fuse to enable repair. SOX7, a member of the SRY-related HMG-box family of transcription factors is expressed in quiescent satellite cells. To elucidate SOX7 function in skeletal muscle, we knocked down Sox7 expression in embryonic stem cells and primary myoblasts and generated a conditional knockout mouse in which Sox7 is excised in PAX3+ cells. Loss of Sox7 in embryonic stem cells reduced Pax3 and Pax7 expression. In vivo, conditional knockdown of Sox7 reduced the satellite cell population from birth, reduced myofiber caliber, and impaired regeneration after acute injury. Although Sox7-deficient primary myoblasts differentiated normally, impaired myoblast fusion and increased sensitivity to apoptosis in culture and in vivo were observed. Taken together, these results indicate that SOX7 is dispensable for myogenesis but is necessary to promote satellite cell development and survival.
Collapse
Affiliation(s)
- Rashida F Rajgara
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Neena Lala-Tabbert
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - François Marchildon
- Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Émilie Lamarche
- Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jennifer K MacDonald
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandre Blais
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ilona S Skerjanc
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nadine Wiper-Bergeron
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
17
|
Kanki Y, Nakaki R, Shimamura T, Matsunaga T, Yamamizu K, Katayama S, Suehiro JI, Osawa T, Aburatani H, Kodama T, Wada Y, Yamashita JK, Minami T. Dynamically and epigenetically coordinated GATA/ETS/SOX transcription factor expression is indispensable for endothelial cell differentiation. Nucleic Acids Res 2017; 45:4344-4358. [PMID: 28334937 PMCID: PMC5416769 DOI: 10.1093/nar/gkx159] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 02/25/2017] [Indexed: 12/29/2022] Open
Abstract
Although studies of the differentiation from mouse embryonic stem (ES) cells to vascular endothelial cells (ECs) provide an excellent model for investigating the molecular mechanisms underlying vascular development, temporal dynamics of gene expression and chromatin modifications have not been well studied. Herein, using transcriptomic and epigenomic analyses based on H3K4me3 and H3K27me3 modifications at a genome-wide scale, we analysed the EC differentiation steps from ES cells and crucial epigenetic modifications unique to ECs. We determined that Gata2, Fli1, Sox7 and Sox18 are master regulators of EC that are induced following expression of the haemangioblast commitment pioneer factor, Etv2. These master regulator gene loci were repressed by H3K27me3 throughout the mesoderm period but rapidly transitioned to histone modification switching from H3K27me3 to H3K4me3 after treatment with vascular endothelial growth factor. SiRNA knockdown experiments indicated that these regulators are indispensable not only for proper EC differentiation but also for blocking the commitment to other closely aligned lineages. Collectively, our detailed epigenetic analysis may provide an advanced model for understanding temporal regulation of chromatin signatures and resulting gene expression profiles during EC commitment. These studies may inform the future development of methods to stimulate the vascular endothelium for regenerative medicine.
Collapse
Affiliation(s)
- Yasuharu Kanki
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan.,Division of Vascular Biology, RCAST, The University of Tokyo, Tokyo 153-8904, Japan.,Division of Systems Biology, RCAST, The University of Tokyo, Tokyo 153-8904, Japan
| | - Ryo Nakaki
- Division of Genome Sciences, RCAST, The University of Tokyo, Tokyo 153-8904, Japan
| | - Teppei Shimamura
- Department of Systems Biology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Taichi Matsunaga
- Department of Stem Cell Differentiation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Deparment of Cell Growth and Differentiation, CiRA, Kyoto University, Kyoto 606-8507, Japan
| | - Kohei Yamamizu
- Department of Stem Cell Differentiation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Deparment of Cell Growth and Differentiation, CiRA, Kyoto University, Kyoto 606-8507, Japan
| | - Shiori Katayama
- Department of Stem Cell Differentiation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Deparment of Cell Growth and Differentiation, CiRA, Kyoto University, Kyoto 606-8507, Japan
| | - Jun-Ichi Suehiro
- Division of Vascular Biology, RCAST, The University of Tokyo, Tokyo 153-8904, Japan.,Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Tokyo 181-8611, Japan
| | - Tsuyoshi Osawa
- Division of Vascular Biology, RCAST, The University of Tokyo, Tokyo 153-8904, Japan.,Division of Systems Biology, RCAST, The University of Tokyo, Tokyo 153-8904, Japan
| | - Hiroyuki Aburatani
- Division of Genome Sciences, RCAST, The University of Tokyo, Tokyo 153-8904, Japan
| | - Tatsuhiko Kodama
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan.,Division of Systems Biology, RCAST, The University of Tokyo, Tokyo 153-8904, Japan
| | - Youichiro Wada
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Jun K Yamashita
- Department of Stem Cell Differentiation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Deparment of Cell Growth and Differentiation, CiRA, Kyoto University, Kyoto 606-8507, Japan
| | - Takashi Minami
- Division of Vascular Biology, RCAST, The University of Tokyo, Tokyo 153-8904, Japan.,Division of Molecular and Vascular Biology, IRDA, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
18
|
Lilly AJ, Mazan A, Scott DA, Lacaud G, Kouskoff V. SOX7 expression is critically required in FLK1-expressing cells for vasculogenesis and angiogenesis during mouse embryonic development. Mech Dev 2017; 146:31-41. [PMID: 28577909 PMCID: PMC5496588 DOI: 10.1016/j.mod.2017.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/27/2017] [Accepted: 05/31/2017] [Indexed: 01/01/2023]
Abstract
The transcriptional program that regulates the differentiation of endothelial precursor cells into a highly organized vascular network is still poorly understood. Here we explore the role of SOX7 during this process, performing a detailed analysis of the vascular defects resulting from either a complete deficiency in Sox7 expression or from the conditional deletion of Sox7 in FLK1-expressing cells. We analysed the consequence of Sox7 deficiency from E7.5 onward to determine from which stage of development the effect of Sox7 deficiency can be observed. We show that while Sox7 is expressed at the onset of endothelial specification from mesoderm, Sox7 deficiency does not impact the emergence of the first endothelial progenitors. However, by E8.5, clear signs of defective vascular development are already observed with the presence of highly unorganised endothelial cords rather than distinct paired dorsal aorta. By E10.5, both Sox7 complete knockout and FLK1-specific deletion of Sox7 lead to widespread vascular defects. In contrast, while SOX7 is expressed in the earliest specified blood progenitors, the VAV-specific deletion of Sox7 does not affect the hematopoietic system. Together, our data reveal the unique role of SOX7 in vasculogenesis and angiogenesis during embryonic development.
Collapse
Affiliation(s)
- Andrew J Lilly
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow road, M20 4BX, UK
| | - Andrzej Mazan
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow road, M20 4BX, UK
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, BCM227, Houston, TX 77030, USA
| | - Georges Lacaud
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow road, M20 4BX, UK.
| | - Valerie Kouskoff
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow road, M20 4BX, UK; Division of Developmental Biology and Medicine, The University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
19
|
Pickering J, Cunliffe VT, Van Eeden F, Borycki AG. Hedgehog signalling acts upstream of Laminin alpha1 transcription in the zebrafish paraxial mesoderm. Matrix Biol 2016; 62:58-74. [PMID: 27856309 DOI: 10.1016/j.matbio.2016.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 12/01/2022]
Abstract
Laminin-111 (α1β1γ1) is a member of the Laminin family of extra-cellular matrix proteins that comprises 16 members, components of basement membranes. Laminin-111, one of the first Laminin proteins synthesised during embryogenesis, is required for basement membrane deposition and has essential roles in tissue morphogenesis and patterning. Yet, the mechanisms controlling Laminin-111 expression are poorly understood. We generated a zebrafish transgenic reporter line that reproduces faithfully the expression pattern of lama1, the gene encoding Laminin α1, and we used this reporter line to investigate lama1 transcriptional regulation. Our findings established that lama1 expression is controlled by intronic enhancers, including an enhancer directing expression in the paraxial mesoderm, anterior spinal cord and hindbrain, located in intron 1. We show that Hedgehog signalling is necessary and sufficient for lama1 transcription in the paraxial mesoderm and identify putative Gli/Zic binding sites that may mediate this control. These findings uncover a conserved role for Hedgehog signalling in the control of basement membrane assembly via its transcriptional regulation of lama1, and provide a mechanism to coordinate muscle cell fate specification in the zebrafish embryo.
Collapse
Affiliation(s)
- Joseph Pickering
- Bateson Centre, Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Vincent T Cunliffe
- Bateson Centre, Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Freek Van Eeden
- Bateson Centre, Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Anne-Gaëlle Borycki
- Bateson Centre, Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK.
| |
Collapse
|
20
|
NF-Y in invertebrates. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1860:630-635. [PMID: 27793714 DOI: 10.1016/j.bbagrm.2016.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 01/07/2023]
Abstract
Both Drosophila melanogaster and Caenorhabditis elegans (C. elegans) are useful model organisms to study in vivo roles of NF-Y during development. Drosophila NF-Y (dNF-Y) consists of three subunits dNF-YA, dNF-YB and dNF-YC. In some tissues, dNF-YC-related protein Mes4 may replace dNF-YC in dNF-Y complex. Studies with eye imaginal disc-specific dNF-Y-knockdown flies revealed that dNF-Y positively regulates the sevenless gene encoding a receptor tyrosine kinase, a component of the ERK pathway and negatively regulates the Sensless gene encoding a transcription factor to ensure proper development of R7 photoreceptor cells together with proper R7 axon targeting. dNF-Y also controls the Drosophila Bcl-2 (debcl) to regulate apoptosis. In thorax development, dNF-Y is necessary for both proper Drosophila JNK (basket) expression and JNK signaling activity that is responsible for thorax development. Drosophila p53 gene was also identified as one of the dNF-Y target genes in this system. C. elegans contains two forms of NF-YA subunit, CeNF-YA1 and CeNF-YA2. C. elegans NF-Y (CeNF-Y) therefore consists of CeNF-YB, CeNF-YC and either CeNF-YA1 or CeNF-YA2. CeNF-Y negatively regulates expression of the Hox gene egl-5 (ortholog of Drosophila Abdominal-B) that is involved in tail patterning. CeNF-Y also negatively regulates expression of the tbx-2 gene that is essential for development of the pharyngeal muscles, specification of neural cell fate and adaptation in olfactory neurons. Negative regulation of the expression of egl-5 and tbx-2 by CeNF-Y provides new insight into the physiological meaning of negative regulation of gene expression by NF-Y during development. In addition, studies on NF-Y in platyhelminths are also summarized. This article is part of a Special Issue entitled: Nuclear Factor Y in Development and Disease, edited by Prof. Roberto Mantovani.
Collapse
|
21
|
Cuvertino S, Lacaud G, Kouskoff V. SOX7-enforced expression promotes the expansion of adult blood progenitors and blocks B-cell development. Open Biol 2016; 6:160070. [PMID: 27411892 PMCID: PMC4967825 DOI: 10.1098/rsob.160070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/22/2016] [Indexed: 12/29/2022] Open
Abstract
During embryogenesis, the three SOXF transcription factors, SOX7, SOX17 and SOX18, regulate the specification of the cardiovascular system and are also involved in the development of haematopoiesis. The ectopic expression of SOX17 in both embryonic and adult blood cells enhances self-renewal. Likewise, the enforced expression of SOX7 during embryonic development promotes the proliferation of early blood progenitors and blocks lineage commitment. However, whether SOX7 expression can also affect the self-renewal of adult blood progenitors has never been explored. In this study, we demonstrate using an inducible transgenic mouse model that the enforced expression of Sox7 ex vivo in bone marrow/stroma cell co-culture promotes the proliferation of blood progenitors which retain multi-lineage short-term engrafting capacity. Furthermore, SOX7 expression induces a profound block in the generation of B lymphocytes. Correspondingly, the ectopic expression of SOX7 in vivo results in dramatic alterations of the haematopoietic system, inducing the proliferation of blood progenitors in the bone marrow while blocking B lymphopoiesis. In addition, SOX7 expression induces extra-medullary haematopoiesis in the spleen and liver. Together, these data demonstrate that the uncontrolled expression of the transcription factor SOX7 in adult haematopoietic cells has dramatic consequences on blood homeostasis.
Collapse
Affiliation(s)
- Sara Cuvertino
- Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Georges Lacaud
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Valerie Kouskoff
- Stem Cell Hematopoiesis Group, Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
22
|
Genome-Wide Identification and Transcriptome-Based Expression Profiling of the Sox Gene Family in the Nile Tilapia (Oreochromis niloticus). Int J Mol Sci 2016; 17:270. [PMID: 26907269 PMCID: PMC4813134 DOI: 10.3390/ijms17030270] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/06/2016] [Accepted: 02/15/2016] [Indexed: 11/16/2022] Open
Abstract
The Sox transcription factor family is characterized with the presence of a Sry-related high-mobility group (HMG) box and plays important roles in various biological processes in animals, including sex determination and differentiation, and the development of multiple organs. In this study, 27 Sox genes were identified in the genome of the Nile tilapia (Oreochromis niloticus), and were classified into seven groups. The members of each group of the tilapia Sox genes exhibited a relatively conserved exon-intron structure. Comparative analysis showed that the Sox gene family has undergone an expansion in tilapia and other teleost fishes following their whole genome duplication, and group K only exists in teleosts. Transcriptome-based analysis demonstrated that most of the tilapia Sox genes presented stage-specific and/or sex-dimorphic expressions during gonadal development, and six of the group B Sox genes were specifically expressed in the adult brain. Our results provide a better understanding of gene structure and spatio-temporal expression of the Sox gene family in tilapia, and will be useful for further deciphering the roles of the Sox genes during sex determination and gonadal development in teleosts.
Collapse
|
23
|
Yan HJ, Zhou SY, Li Y, Zhang H, Deng CY, Qi H, Li FR. The effects of LSD1 inhibition on self-renewal and differentiation of human induced pluripotent stem cells. Exp Cell Res 2015; 340:227-37. [PMID: 26748182 DOI: 10.1016/j.yexcr.2015.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 12/25/2015] [Accepted: 12/30/2015] [Indexed: 12/21/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) are capable of unlimited self-renewal and can generate nearly all cells in the body. Changes induced by different LSD1 activities on the regulation of hiPSC self-renewal and differentiation and the mechanism underlying such changes were determined. We used two different LSD1 inhibitors (phenelzine sulfate and tranylcypromine) and RNAi technique to inhibit LSD1 activity, and we obtained hiPSCs showing 71.3%, 53.28%, and 31.33% of the LSD1 activity in normal hiPSCs. The cells still maintained satisfactory self-renewal capacity when LSD1 activity was at 71.3%. The growth rate of hiPSCs decreased and cells differentiated when LSD1 activity was at approximately 53.28%. The hiPSCs were mainly arrested in the G0/G1 phase and simultaneously differentiated into endodermal tissue when LSD1 activity was at 31.33%. Teratoma experiments showed that the downregulation of LSD1 resulted in low teratoma volume. When LSD1 activity was below 50%, pluripotency of hiPSCs was impaired, and the teratomas mainly comprised endodermal and mesodermal tissues. This phenomenon was achieved by regulating the critical balance between histone methylation and demethylation at regulatory regions of several key pluripotent and developmental genes.
Collapse
Affiliation(s)
- Hong-Jie Yan
- The Key Laboratory of Stem Cell and Cellular Therapy, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China; Shenzhen Cell Therapy Public Service Platform, Shenzhen 518020, China
| | - Shu-Yan Zhou
- The Key Laboratory of Stem Cell and Cellular Therapy, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China; Shenzhen Cell Therapy Public Service Platform, Shenzhen 518020, China
| | - Yang Li
- The Key Laboratory of Stem Cell and Cellular Therapy, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China; Shenzhen Cell Therapy Public Service Platform, Shenzhen 518020, China
| | - Hui Zhang
- Nevada Cancer Institute, Las Vegas, NV 89135, USA
| | - Chun-Yan Deng
- The Key Laboratory of Stem Cell and Cellular Therapy, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China; Shenzhen Cell Therapy Public Service Platform, Shenzhen 518020, China
| | - Hui Qi
- The Key Laboratory of Stem Cell and Cellular Therapy, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China; Shenzhen Cell Therapy Public Service Platform, Shenzhen 518020, China
| | - Fu-Rong Li
- The Key Laboratory of Stem Cell and Cellular Therapy, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen 518020, China; Shenzhen Cell Therapy Public Service Platform, Shenzhen 518020, China.
| |
Collapse
|
24
|
Kinoshita M, Shimosato D, Yamane M, Niwa H. Sox7 is dispensable for primitive endoderm differentiation from mouse ES cells. BMC DEVELOPMENTAL BIOLOGY 2015; 15:37. [PMID: 26475439 PMCID: PMC4609079 DOI: 10.1186/s12861-015-0079-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/07/2015] [Indexed: 11/10/2022]
Abstract
Background Primitive endoderm is a cell lineage segregated from the epiblast in the blastocyst and gives rise to parietal and visceral endoderm. Sox7 is a member of the SoxF gene family that is specifically expressed in primitive endoderm in the late blastocyst, although its function in this cell lineage remains unclear. Results Here we characterize the function of Sox7 in primitive endoderm differentiation using mouse embryonic stem (ES) cells as a model system. We show that ectopic expression of Sox7 in ES cells has a marginal effect on triggering differentiation into primitive endoderm-like cells. We also show that targeted disruption of Sox7 in ES cells does not affect differentiation into primitive endoderm cells in embryoid body formation as well as by forced expression of Gata6. Conclusions These data indicate that Sox7 function is supplementary and not essential for this differentiation from ES cells. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0079-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masaki Kinoshita
- Laboratory for Pluripotent cell studies, RIKEN, Centre for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Daisuke Shimosato
- Laboratory for Pluripotent cell studies, RIKEN, Centre for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan. .,Laboratory for Development and Regenerative Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunokicho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Mariko Yamane
- Laboratory for Pluripotent cell studies, RIKEN, Centre for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Hitoshi Niwa
- Laboratory for Pluripotent cell studies, RIKEN, Centre for Developmental Biology, 2-2-3, Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan. .,Laboratory for Development and Regenerative Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunokicho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan. .,Present address: Department of Pluripotent Stem Cell Biology, Institure of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan.
| |
Collapse
|
25
|
She ZY, Yang WX. SOX family transcription factors involved in diverse cellular events during development. Eur J Cell Biol 2015; 94:547-63. [PMID: 26340821 DOI: 10.1016/j.ejcb.2015.08.002] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 08/11/2015] [Accepted: 08/11/2015] [Indexed: 12/22/2022] Open
Abstract
In metazoa, SOX family transcription factors play many diverse roles. In vertebrate, they are well-known regulators of numerous developmental processes. Wide-ranging studies have demonstrated the co-expression of SOX proteins in various developing tissues and that they occur in an overlapping manner and show functional redundancy. In particular, studies focusing on the HMG box of SOX proteins have revealed that the HMG box regulates DNA-binding properties, and mediates both the nucleocytoplasmic shuttling of SOX proteins and their physical interactions with partner proteins. Posttranslational modifications are further implicated in the regulation of the transcriptional activities of SOX proteins. In this review, we discuss the underlying molecular mechanisms involved in the SOX-partner factor interactions and the functional modes of SOX-partner complexes during development. We particularly emphasize the representative roles of the SOX group proteins in major tissues during developmental and physiological processes.
Collapse
Affiliation(s)
- Zhen-Yu She
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China.
| |
Collapse
|
26
|
Tsai PH, Chien Y, Chuang JH, Chou SJ, Chien CH, Lai YH, Li HY, Ko YL, Chang YL, Wang CY, Liu YY, Lee HC, Yang CH, Tsai TF, Lee YY, Chiou SH. Dysregulation of Mitochondrial Functions and Osteogenic Differentiation in Cisd2-Deficient Murine Induced Pluripotent Stem Cells. Stem Cells Dev 2015; 24:2561-76. [PMID: 26230298 DOI: 10.1089/scd.2015.0066] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Wolfram syndrome 2 (WFS2) is a premature aging syndrome caused by an irreversible mitochondria-mediated disorder. Cisd2, which regulates mitochondrial electron transport, has been recently identified as the causative gene of WFS2. The mouse Cisd2 knockout (KO) (Cisd2(-/-)) recapitulates most of the clinical manifestations of WFS2, including growth retardation, osteopenia, and lordokyphosis. However, the precise mechanisms underlying osteopenia in WFS2 and Cisd2 KO mice remain unknown. In this study, we collected embryonic fibroblasts from Cisd2-deficient embryos and reprogrammed them into induced pluripotent stem cells (iPSCs) via retroviral transduction with Oct4/Sox2/Klf4/c-Myc. Cisd2-deficient mouse iPSCs (miPSCs) exhibited structural abnormalities in their mitochondria and an impaired proliferative capability. The global gene expression profiles of Cisd2(+/+), Cisd2(+/-), and Cisd2(-/-) miPSCs revealed that Cisd2 functions as a regulator of both mitochondrial electron transport and Wnt/β-catenin signaling, which is critical for cell proliferation and osteogenic differentiation. Notably, Cisd2(-/-) miPSCs exhibited impaired Wnt/β-catenin signaling, with the downregulation of downstream genes, such as Tcf1, Fosl1, and Jun and the osteogenic regulator Runx2. Several differentiation markers for tridermal lineages were globally impaired in Cisd2(-/-) miPSCs. Alizarin red S staining and flow cytometry analysis further revealed that Cisd2(-/-) miPSCs failed to undergo osteogenic differentiation. Taken together, our results, as determined using an miPSC-based platform, have demonstrated that Cisd2 regulates mitochondrial function, proliferation, intracellular Ca(2+) homeostasis, and Wnt pathway signaling. Cisd2 deficiency impairs the activation of Wnt/β-catenin signaling and thereby contributes to the pathogeneses of osteopenia and lordokyphosis in WFS2 patients.
Collapse
Affiliation(s)
- Ping-Hsing Tsai
- 1 Institute of Pharmacology, National Yang-Ming University , Taipei, Taiwan
| | - Yueh Chien
- 1 Institute of Pharmacology, National Yang-Ming University , Taipei, Taiwan .,2 Department of Medical Research, Taipei Veterans General Hospital , Taipei, Taiwan
| | - Jen-Hua Chuang
- 2 Department of Medical Research, Taipei Veterans General Hospital , Taipei, Taiwan .,3 Institute of Clinical Medicine, National Yang-Ming University , Taipei, Taiwan
| | - Shih-Jie Chou
- 1 Institute of Pharmacology, National Yang-Ming University , Taipei, Taiwan
| | - Chian-Hsu Chien
- 2 Department of Medical Research, Taipei Veterans General Hospital , Taipei, Taiwan .,3 Institute of Clinical Medicine, National Yang-Ming University , Taipei, Taiwan
| | - Ying-Hsiu Lai
- 4 Institute of Anatomy & Cell Biology, National Yang-Ming University , Taipei, Taiwan
| | - Hsin-Yang Li
- 4 Institute of Anatomy & Cell Biology, National Yang-Ming University , Taipei, Taiwan .,5 School of Medicine, National Yang-Ming University , Taipei, Taiwan .,6 Department of Obstetrics and Gynecology, Neurological Institute , Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Lin Ko
- 2 Department of Medical Research, Taipei Veterans General Hospital , Taipei, Taiwan .,5 School of Medicine, National Yang-Ming University , Taipei, Taiwan
| | - Yuh-Lih Chang
- 1 Institute of Pharmacology, National Yang-Ming University , Taipei, Taiwan .,7 Department of Pharmacy, Neurological Institute , Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chen-Ying Wang
- 5 School of Medicine, National Yang-Ming University , Taipei, Taiwan
| | - Yung-Yang Liu
- 2 Department of Medical Research, Taipei Veterans General Hospital , Taipei, Taiwan .,3 Institute of Clinical Medicine, National Yang-Ming University , Taipei, Taiwan
| | - Hsin-Chen Lee
- 1 Institute of Pharmacology, National Yang-Ming University , Taipei, Taiwan .,5 School of Medicine, National Yang-Ming University , Taipei, Taiwan
| | - Chang-Hao Yang
- 8 Department of Ophthalmology, National Taiwan University Hospital , Taipei, Taiwan
| | - Ting-Fen Tsai
- 9 Department of Life Sciences & Institute of Genome Sciences, National Yang-Ming University , Taipei, Taiwan
| | - Yi-Yen Lee
- 3 Institute of Clinical Medicine, National Yang-Ming University , Taipei, Taiwan .,10 Department of Neurosurgery, Neurological Institute , Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Hwa Chiou
- 1 Institute of Pharmacology, National Yang-Ming University , Taipei, Taiwan .,2 Department of Medical Research, Taipei Veterans General Hospital , Taipei, Taiwan .,3 Institute of Clinical Medicine, National Yang-Ming University , Taipei, Taiwan .,4 Institute of Anatomy & Cell Biology, National Yang-Ming University , Taipei, Taiwan
| |
Collapse
|
27
|
Viotti M, Foley AC, Hadjantonakis AK. Gutsy moves in mice: cellular and molecular dynamics of endoderm morphogenesis. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0547. [PMID: 25349455 DOI: 10.1098/rstb.2013.0547] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite the importance of the gut and its accessory organs, our understanding of early endoderm development is still incomplete. Traditionally, endoderm has been difficult to study because of its small size and relative fragility. However, recent advances in live cell imaging technologies have dramatically expanded our understanding of this tissue, adding a new appreciation for the complex molecular and morphogenetic processes that mediate gut formation. Several spatially and molecularly distinct subpopulations have been shown to exist within the endoderm before the onset of gastrulation. Here, we review findings that have uncovered complex cell movements within the endodermal layer, before and during gastrulation, leading to the conclusion that cells from primitive endoderm contribute descendants directly to gut.
Collapse
Affiliation(s)
- Manuel Viotti
- Genentech Incorporated, South San Francisco, CA 94080, USA
| | - Ann C Foley
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA
| | | |
Collapse
|
28
|
Robinson AS, Materna SC, Barnes RM, De Val S, Xu SM, Black BL. An arterial-specific enhancer of the human endothelin converting enzyme 1 (ECE1) gene is synergistically activated by Sox17, FoxC2, and Etv2. Dev Biol 2014; 395:379-389. [PMID: 25179465 DOI: 10.1016/j.ydbio.2014.08.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/19/2014] [Indexed: 11/19/2022]
Abstract
Endothelin-converting enzyme-1 (Ece-1), a crucial component of the Endothelin signaling pathway, is required for embryonic development and is an important regulator of vascular tone, yet the transcriptional regulation of the ECE1 gene has remained largely unknown. Here, we define the activity and regulation of an enhancer from the human ECE1 locus in vivo. The enhancer identified here becomes active in endothelial progenitor cells shortly after their initial specification and is dependent on a conserved FOX:ETS motif, a composite binding site for Forkhead transcription factors and the Ets transcription factor Etv2, for activity in vivo. The ECE1 FOX:ETS motif is bound and cooperatively activated by FoxC2 and Etv2, but unlike other described FOX:ETS-dependent enhancers, ECE1 enhancer activity becomes restricted to arterial endothelium and endocardium by embryonic day 9.5 in transgenic mouse embryos. The ECE1 endothelial enhancer also contains an evolutionarily-conserved, consensus SOX binding site, which is required for activity in transgenic mouse embryos. Importantly, the ECE1 SOX site is bound and activated by Sox17, a transcription factor involved in endothelial cell differentiation and an important regulator of arterial identity. Moreover, the ECE1 enhancer is cooperatively activated by the combinatorial action of FoxC2, Etv2, and Sox17. Although Sox17 is required for arterial identity, few direct transcriptional targets have been identified in endothelial cells. Thus, this work has important implications for our understanding of endothelial specification and arterial subspecification.
Collapse
Affiliation(s)
- Ashley S Robinson
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158-2517
| | - Stefan C Materna
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158-2517
| | - Ralston M Barnes
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158-2517
| | - Sarah De Val
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158-2517
| | - Shan-Mei Xu
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158-2517
| | - Brian L Black
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158-2517
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158-2517
| |
Collapse
|
29
|
Aksoy I, Giudice V, Delahaye E, Wianny F, Aubry M, Mure M, Chen J, Jauch R, Bogu GK, Nolden T, Himmelbauer H, Xavier Doss M, Sachinidis A, Schulz H, Hummel O, Martinelli P, Hübner N, Stanton LW, Real FX, Bourillot PY, Savatier P. Klf4 and Klf5 differentially inhibit mesoderm and endoderm differentiation in embryonic stem cells. Nat Commun 2014; 5:3719. [PMID: 24770696 DOI: 10.1038/ncomms4719] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 03/24/2014] [Indexed: 01/04/2023] Open
Abstract
Krüppel-like factors (Klf) 4 and 5 are two closely related members of the Klf family, known to play key roles in cell cycle regulation, somatic cell reprogramming and pluripotency. Here we focus on the functional divergence between Klf4 and Klf5 in the inhibition of mouse embryonic stem (ES) cell differentiation. Using microarrays and chromatin immunoprecipitation coupled to ultra-high-throughput DNA sequencing, we show that Klf4 negatively regulates the expression of endodermal markers in the undifferentiated ES cells, including transcription factors involved in the commitment of pluripotent stem cells to endoderm differentiation. Knockdown of Klf4 enhances differentiation towards visceral and definitive endoderm. In contrast, Klf5 negatively regulates the expression of mesodermal markers, some of which control commitment to the mesoderm lineage, and knockdown of Klf5 specifically enhances differentiation towards mesoderm. We conclude that Klf4 and Klf5 differentially inhibit mesoderm and endoderm differentiation in murine ES cells.
Collapse
Affiliation(s)
- Irène Aksoy
- 1] Inserm, U846, 18 Avenue Doyen Lepine, Bron 69500, France [2] Stem Cell and Brain Research Institute, Bron 69500, France [3] Université de Lyon, Université Lyon 1, Lyon 69003, France [4] Genome Institute of Singapore, 60 Biopolis street, Singapore 138672, Singapore [5]
| | - Vincent Giudice
- 1] Inserm, U846, 18 Avenue Doyen Lepine, Bron 69500, France [2] Stem Cell and Brain Research Institute, Bron 69500, France [3] Université de Lyon, Université Lyon 1, Lyon 69003, France [4]
| | - Edwige Delahaye
- 1] Inserm, U846, 18 Avenue Doyen Lepine, Bron 69500, France [2] Stem Cell and Brain Research Institute, Bron 69500, France [3] Université de Lyon, Université Lyon 1, Lyon 69003, France
| | - Florence Wianny
- 1] Inserm, U846, 18 Avenue Doyen Lepine, Bron 69500, France [2] Stem Cell and Brain Research Institute, Bron 69500, France [3] Université de Lyon, Université Lyon 1, Lyon 69003, France
| | - Maxime Aubry
- 1] Inserm, U846, 18 Avenue Doyen Lepine, Bron 69500, France [2] Stem Cell and Brain Research Institute, Bron 69500, France [3] Université de Lyon, Université Lyon 1, Lyon 69003, France
| | - Magali Mure
- 1] Inserm, U846, 18 Avenue Doyen Lepine, Bron 69500, France [2] Stem Cell and Brain Research Institute, Bron 69500, France [3] Université de Lyon, Université Lyon 1, Lyon 69003, France
| | - Jiaxuan Chen
- Genome Institute of Singapore, 60 Biopolis street, Singapore 138672, Singapore
| | - Ralf Jauch
- 1] Genome Institute of Singapore, 60 Biopolis street, Singapore 138672, Singapore [2] Genome Regulation Laboratory, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Gireesh K Bogu
- Genome Institute of Singapore, 60 Biopolis street, Singapore 138672, Singapore
| | - Tobias Nolden
- Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany
| | - Heinz Himmelbauer
- 1] Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany [2] Center for Genomic Regulation (CRG), C. Dr. Aiguader 88, Barcelona 08003, Spain [3] Universitat Pompeu Fabra (UPF), C. Dr. Aiguader 88, Barcelona 08003, Spain
| | - Michael Xavier Doss
- 1] Universitat Pompeu Fabra (UPF), C. Dr. Aiguader 88, Barcelona 08003, Spain [2]
| | - Agapios Sachinidis
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, Robert-Koch-Strasse. 39, Cologne 50931, Germany
| | - Herbert Schulz
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, Berlin 13125, Germany
| | - Oliver Hummel
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, Berlin 13125, Germany
| | - Paola Martinelli
- Centro Nacional de Investigaciones Oncológicas, Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, Berlin 13125, Germany
| | - Lawrence W Stanton
- Genome Institute of Singapore, 60 Biopolis street, Singapore 138672, Singapore
| | - Francisco X Real
- 1] Centro Nacional de Investigaciones Oncológicas, Melchor Fernández Almagro 3, Madrid 28029, Spain [2] Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Pierre-Yves Bourillot
- 1] Inserm, U846, 18 Avenue Doyen Lepine, Bron 69500, France [2] Stem Cell and Brain Research Institute, Bron 69500, France [3] Université de Lyon, Université Lyon 1, Lyon 69003, France
| | - Pierre Savatier
- 1] Inserm, U846, 18 Avenue Doyen Lepine, Bron 69500, France [2] Stem Cell and Brain Research Institute, Bron 69500, France [3] Université de Lyon, Université Lyon 1, Lyon 69003, France
| |
Collapse
|
30
|
Stovall DB, Cao P, Sui G. SOX7: from a developmental regulator to an emerging tumor suppressor. Histol Histopathol 2013; 29:439-45. [PMID: 24288056 DOI: 10.14670/hh-29.10.439] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
SOX7 belongs to the SOX (SRY-related HMG-box) family of transcription factors that have been shown to regulate multiple biological processes, such as hematopoiesis, vasculogenesis and cardiogenesis during embryonic development. Recent studies indicate that several SOX family members play important roles in tumorigenesis. In this review, we introduce SOX7 gene and protein structures, and discuss its expression and functional role in cancer development and progression. SOX7 is frequently downregulated in many human cancers and its reduced expression correlates with poor prognoses of several cancers. Functional studies reveal many tumor suppressive properties of SOX7 in prostate, colon, lung, and breast cancers. To date, although a few target genes of SOX7 have been identified, SOX7-mediated gene expression has not been investigated in a cancer-relevant context. Our recent studies not only for the first time demonstrate a tumor suppressive role of SOX7 in a xenograft mouse model, but also unravel that many genes regulating cell death, growth and apoptosis are affected by SOX7, strongly supporting a pivotal role of SOX7 in tumorigenesis. Thus, currently available data clearly indicate a tumor suppressive role of SOX7, but the mechanisms underlying its gene expression and tumor suppressive activity remain undetermined. The research of SOX7 in cancers remains a fertile area to be explored.
Collapse
Affiliation(s)
- Daniel B Stovall
- Department of Cancer Biology and Comprehensive Cancer Center, and Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Paul Cao
- Department of Cancer Biology and Comprehensive Cancer Center, and Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Guangchao Sui
- Department of Cancer Biology and Comprehensive Cancer Center, and Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
31
|
The regulation of SOX7 and its tumor suppressive role in breast cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1645-1653. [PMID: 24012678 DOI: 10.1016/j.ajpath.2013.07.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/16/2013] [Accepted: 07/10/2013] [Indexed: 12/23/2022]
Abstract
Both epigenetic silencing and genetic deletion of tumor suppressors contribute to the development and progression of breast cancer. SOX7 is a transcription factor important to development, and its down-regulation has been reported in tumor tissues and cell lines of prostate, colon, and lung cancers. However, the regulation of SOX7 expression and its functional role in breast cancer have not been reported. The current study demonstrates that SOX7 mRNA and protein expression are down-regulated in breast cancer tissues and cell lines compared with adjacent normal tissues and nontumorigenic cells, respectively. The SOX7 promoter is hypermethylated in breast cancer cell lines compared with nontumorigenic cells, and the inhibition of DNA methylation increases SOX7 mRNA levels. With shRNA-mediated SOX7 silencing, nontumorigenic immortal breast cells display increased proliferation, migration, and invasion and form structures that resemble that of breast cancer cells in a three-dimensional culture system. Conversely, ectopic SOX7 expression inhibits proliferation, migration, and invasion of breast cancer cells in vitro and tumor growth in vivo. Importantly, we discovered that SOX7 transcript levels positively correlated with clinical outcome of 674 breast cancer patients. Overall, our data suggest that SOX7 acts as a tumor suppressor in breast cancer. SOX7 expression is likely regulated by multiple mechanisms and potentially serves as a prognostic marker for breast cancer patients.
Collapse
|
32
|
Ly LL, Suyari O, Yoshioka Y, Tue NT, Yoshida H, Yamaguchi M. dNF-YB plays dual roles in cell death and cell differentiation during Drosophila eye development. Gene 2013; 520:106-18. [DOI: 10.1016/j.gene.2013.02.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/16/2013] [Accepted: 02/23/2013] [Indexed: 11/16/2022]
|
33
|
Uemura M, Ozawa A, Nagata T, Kurasawa K, Tsunekawa N, Nobuhisa I, Taga T, Hara K, Kudo A, Kawakami H, Saijoh Y, Kurohmaru M, Kanai-Azuma M, Kanai Y. Sox17 haploinsufficiency results in perinatal biliary atresia and hepatitis in C57BL/6 background mice. Development 2013; 140:639-48. [PMID: 23293295 DOI: 10.1242/dev.086702] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Congenital biliary atresia is an incurable disease of newborn infants, of unknown genetic causes, that results in congenital deformation of the gallbladder and biliary duct system. Here, we show that during mouse organogenesis, insufficient SOX17 expression in the gallbladder and bile duct epithelia results in congenital biliary atresia and subsequent acute 'embryonic hepatitis', leading to perinatal death in ~95% of the Sox17 heterozygote neonates in C57BL/6 (B6) background mice. During gallbladder and bile duct development, Sox17 was expressed at the distal edge of the gallbladder primordium. In the Sox17(+/-) B6 embryos, gallbladder epithelia were hypoplastic, and some were detached from the luminal wall, leading to bile duct stenosis or atresia. The shredding of the gallbladder epithelia is probably caused by cell-autonomous defects in proliferation and maintenance of the Sox17(+/-) gallbladder/bile duct epithelia. Our results suggest that Sox17 plays a dosage-dependent function in the morphogenesis and maturation of gallbladder and bile duct epithelia during the late-organogenic stages, highlighting a novel entry point to the understanding of the etiology and pathogenesis of human congenital biliary atresia.
Collapse
Affiliation(s)
- Mami Uemura
- Department of Veterinary Anatomy, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Affiliation(s)
- Anna Domogatskaya
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden; , ,
| | - Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden; , ,
| | - Karl Tryggvason
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden; , ,
| |
Collapse
|
35
|
Feng X, Zhang J, Smuga-Otto K, Tian S, Yu J, Stewart R, Thomson JA. Protein kinase C mediated extraembryonic endoderm differentiation of human embryonic stem cells. Stem Cells 2012; 30:461-70. [PMID: 22213079 DOI: 10.1002/stem.1018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Unlike mouse embryonic stem cells (ESCs), which are closely related to the inner cell mass, human ESCs appear to be more closely related to the later primitive ectoderm. For example, human ESCs and primitive ectoderm share a common epithelial morphology, growth factor requirements, and the potential to differentiate to all three embryonic germ layers. However, it has previously been shown that human ESCs can also differentiate to cells expressing markers of trophoblast, an extraembryonic lineage formed before the formation of primitive ectoderm. Here, we show that phorbol ester 12-O-tetradecanoylphorbol 13-acetate causes human ESCs to undergo an epithelial mesenchymal transition and to differentiate into cells expressing markers of parietal endoderm, another extraembryonic lineage. We further confirmed that this differentiation is through the activation of protein kinase C (PKC) pathway and demonstrated that a particular PKC subtype, PKC-δ, is most responsible for this transition.
Collapse
Affiliation(s)
- Xuezhu Feng
- Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
The crystal structure of the Sox4 HMG domain-DNA complex suggests a mechanism for positional interdependence in DNA recognition. Biochem J 2012; 443:39-47. [PMID: 22181698 DOI: 10.1042/bj20111768] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
It has recently been proposed that the sequence preferences of DNA-binding TFs (transcription factors) can be well described by models that include the positional interdependence of the nucleotides of the target sites. Such binding models allow for multiple motifs to be invoked, such as principal and secondary motifs differing at two or more nucleotide positions. However, the structural mechanisms underlying the accommodation of such variant motifs by TFs remain elusive. In the present study we examine the crystal structure of the HMG (high-mobility group) domain of Sox4 [Sry (sex-determining region on the Y chromosome)-related HMG box 4] bound to DNA. By comparing this structure with previously solved structures of Sox17 and Sox2, we observed subtle conformational differences at the DNA-binding interface. Furthermore, using quantitative electrophoretic mobility-shift assays we validated the positional interdependence of two nucleotides and the presence of a secondary Sox motif in the affinity landscape of Sox4. These results suggest that a concerted rearrangement of two interface amino acids enables Sox4 to accommodate primary and secondary motifs. The structural adaptations lead to altered dinucleotide preferences that mutually reinforce each other. These analyses underline the complexity of the DNA recognition by TFs and provide an experimental validation for the conceptual framework of positional interdependence and secondary binding motifs.
Collapse
|
37
|
Yoshioka Y, Ly LL, Yamaguchi M. Transcription factor NF-Y is involved in differentiation of R7 photoreceptor cell in Drosophila. Biol Open 2011; 1:19-29. [PMID: 23213364 PMCID: PMC3507159 DOI: 10.1242/bio.2011013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The CCAAT motif-binding factor NF-Y consists of three different subunits, NF-YA, NF-YB and NF-YC. Knockdown of Drosophila NF-YA (dNF-YA) in eye discs with GMR-GAL4 and UAS-dNF-YAIR resulted in a rough eye phenotype and monitoring of differentiation of photoreceptor cells by LacZ expression in seven up-LacZ and deadpan-lacZ enhancer trap lines revealed associated loss of R7 photoreceptor signals. In line with differentiation of R7 being regulated by the sevenless (sev) gene and the MAPK cascade, the rough eye phenotype and loss of R7 signals in dNF-YA-knockdown flies were rescued by expression of the sev gene, or the D-raf gene, a downstream component of the MAPK cascade. The sev gene promoter contains two dNF-Y-binding consensus sequences which play positive roles in promoter activity. In chromatin immunoprecipitation assays with anti-dNF-YA antibody and S2 cells, the sev gene promoter region containing the NF-Y consensus was effectively amplified in immunoprecipitates from transgenic flies by polymerase chain reaction, indicating that dNF-Y is necessary for appropriate sev expression and involved in R7 photoreceptor cell development.
Collapse
Affiliation(s)
- Yasuhide Yoshioka
- Department of Applied Biology, Kyoto Institute of Technology , Matsugasaki, Sakyo-ku, Kyoto 606-8585 , Japan ; Venture Laboratory, Kyoto Institute of Technology , Matsugasaki, Sakyo-ku, Kyoto 606-8585 , Japan
| | | | | |
Collapse
|
38
|
Synergic reprogramming of mammalian cells by combined exposure to mitotic Xenopus egg extracts and transcription factors. Proc Natl Acad Sci U S A 2011; 108:17331-6. [PMID: 21908712 DOI: 10.1073/pnas.1100733108] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Transfer of somatic cell nuclei to enucleated eggs and ectopic expression of specific transcription factors are two different reprogramming strategies used to generate pluripotent cells from differentiated cells. However, these methods are poorly efficient, and other unknown factors might be required to increase their success rate. Here we show that Xenopus egg extracts at the metaphase stage (M phase) have a strong reprogramming activity on mouse embryonic fibroblasts (MEFs). First, they reset replication properties of MEF nuclei toward a replication profile characteristic of early development, and they erase several epigenetic marks, such as trimethylation of H3K9, H3K4, and H4K20. Second, when MEFs are reversibly permeabilized in the presence of M-phase Xenopus egg extracts, they show a transient increase in cell proliferation, form colonies, and start to express specific pluripotency markers. Finally, transient exposure of MEF nuclei to M-phase Xenopus egg extracts increases the success of nuclear transfer to enucleated mouse oocytes and strongly synergizes with the production of pluripotent stem cells by ectopic expression of transcription factors. The mitotic stage of the egg extract is crucial, because none of these effects is detected when using interphasic Xenopus egg extracts. Our data demonstrate that mitosis is essential to make mammalian somatic nuclei prone to reprogramming and that, surprisingly, the heterologous Xenopus system has features that are conserved enough to remodel mammalian nuclei.
Collapse
|
39
|
Artus J, Piliszek A, Hadjantonakis AK. The primitive endoderm lineage of the mouse blastocyst: sequential transcription factor activation and regulation of differentiation by Sox17. Dev Biol 2010; 350:393-404. [PMID: 21146513 DOI: 10.1016/j.ydbio.2010.12.007] [Citation(s) in RCA: 172] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 11/30/2010] [Accepted: 12/02/2010] [Indexed: 01/06/2023]
Abstract
Cells of the primitive endoderm (PrE) and the pluripotent epiblast (EPI), the two lineages specified within the inner cell mass (ICM) of the mouse blastocyst stage embryo, are segregated into adjacent tissue layers by the end of the preimplantation period. The PrE layer which emerges as a polarized epithelium adjacent to the blastocoel, with a basement membrane separating it from the EPI, has two derivatives, the visceral and parietal endoderm. In this study we have investigated the localization of two transcriptional regulators of the SOX family, SOX17 and SOX7, within the PrE and its derivatives. We noted that SOX17 was first detected in a salt-and-pepper distribution within the ICM, subsequently becoming restricted to the nascent PrE epithelium. This dynamic distribution of SOX17 resembled the localization of GATA6 and GATA4, two other PrE lineage-specific transcription factors. By contrast, SOX7 was only detected in PrE cells positioned in contact with the blastocoel, raising the possibility that these cells are molecularly distinct. Our observations support a model of sequential GATA6 > SOX17 > GATA4 > SOX7 transcription factor activation within the PrE lineage, perhaps correlating with the consecutive periods of cell lineage 'naïvete', commitment and sorting. Furthermore our data suggest that co-expression of SOX17 and SOX7 within sorted PrE cells could account for the absence of a detectable phenotype of Sox17 mutant blastocysts. However, analysis of implantation-delayed blastocysts, revealed a role for SOX17 in the maintenance of PrE epithelial integrity, with the absence of SOX17 leading to premature delamination and migration of parietal endoderm.
Collapse
Affiliation(s)
- Jérôme Artus
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | | | | |
Collapse
|
40
|
Mauney JR, Ramachandran A, Yu RN, Daley GQ, Adam RM, Estrada CR. All-trans retinoic acid directs urothelial specification of murine embryonic stem cells via GATA4/6 signaling mechanisms. PLoS One 2010; 5:e11513. [PMID: 20644631 PMCID: PMC2903484 DOI: 10.1371/journal.pone.0011513] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 06/02/2010] [Indexed: 01/05/2023] Open
Abstract
The urinary bladder and associated tract are lined by the urothelium, a transitional epithelium that acts as a specialized permeability barrier that protects the underlying tissue from urine via expression of a highly specific group of proteins known as the uroplakins (UP). To date, our understanding of the developmental processes responsible for urothelial differentiation has been hampered due to the lack of suitable models. In this study, we describe a novel in vitro cell culture system for derivation of urothelial cells from murine embryonic stem cells (ESCs) following cultivation on collagen matrices in the presence all trans retinoic acid (RA). Upon stimulation with micromolar concentrations of RA, ESCs significantly downregulated the pluripotency factor OCT-4 but markedly upregulated UP1A, UP1B, UP2, UP3A, and UP3B mRNA levels in comparison to naïve ESCs and spontaneously differentiating controls. Pan-UP protein expression was associated with both p63- and cytokeratin 20-positive cells in discrete aggregating populations of ESCs following 9 and 14 days of RA stimulation. Analysis of endodermal transcription factors such as GATA4 and GATA6 revealed significant upregulation and nuclear enrichment in RA-treated UP2-GFP+ populations. GATA4-/- and GATA6-/- transgenic ESC lines revealed substantial attenuation of RA-mediated UP expression in comparison to wild type controls. In addition, EMSA analysis revealed that RA treatment induced formation of transcriptional complexes containing GATA4/6 on both UP1B and UP2 promoter fragments containing putative GATA factor binding sites. Collectively, these data suggest that RA mediates ESC specification toward a urothelial lineage via GATA4/6-dependent processes.
Collapse
Affiliation(s)
- Joshua R. Mauney
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aruna Ramachandran
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Richard N. Yu
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - George Q. Daley
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Boston, Massachusetts, United States of America
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
- Division of Hematology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Manton Center for Orphan Disease Research, Boston, Massachusetts, United States of America
| | - Rosalyn M. Adam
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Carlos R. Estrada
- Urological Diseases Research Center, Children's Hospital Boston, Boston, Massachusetts, United States of America
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
41
|
Chew LJ, Gallo V. The Yin and Yang of Sox proteins: Activation and repression in development and disease. J Neurosci Res 2010; 87:3277-87. [PMID: 19437544 DOI: 10.1002/jnr.22128] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The general view of development consists of the acquisition of committed/differentiated phenotypes following a period of self-renewal and progenitor expansion. Lineage specification and progression are phenomena of antagonistic events, silencing tissue-specific gene expression in precursors to allow self-renewal and multipotentiality, and subsequently suppressing proliferation and embryonic gene expression to promote the restricted expression of tissue-specific genes during maturation. The high mobility group-containing Sox family of transcription factors constitutes one of the earliest classes of genes to be expressed during embryonic development. These proteins not only are indispensable for progenitor cell specification but also are critical for terminal differentiation of multiple cell types in a wide variety of lineages. Sox transcription factors are now known to induce or repress progenitor cell characteristics and cell proliferation or to activate the expression of tissue-specific genes. Sox proteins fulfill their diverse functions in developmental regulation by distinct molecular mechanisms. Not surprisingly, in addition to DNA binding and bending, Sox transcription factors also interact with different protein partners to function as coactivators or corepressors of downstream target genes. Here we seek to provide an overview of the current knowledge of Sox gene functional mechanisms, in an effort to understand their roles in both development and pathology.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, USA
| | | |
Collapse
|
42
|
The role of the basement membrane as a modulator of intestinal epithelial-mesenchymal interactions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 96:175-206. [PMID: 21075345 DOI: 10.1016/b978-0-12-381280-3.00008-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intestinal development is a process of continuous dynamic bidirectional crosstalk between epithelial and underlying mesenchymal cells. This crosstalk is mediated by well-dissected signaling pathways. Another crucial actor in the epithelio-mesenchymal interactions is the stromal microenvironment, which is composed of extracellular matrix molecules. Among them, the basement membrane (BM) molecules are secreted by the epithelium and mesenchyme in a complementary manner. These molecules signal back to the cells via the integrins or other specific receptors. In this review, we mainly focus on the BM molecules, particularly laminins. The major BM molecules are organized in a complex molecular network, which is highly variable among organs. Cell culture, coculture, and grafting models have been of great interest in understanding the importance of these molecules. Mouse gene ablation of laminin chains are interesting models, which often lead to embryonic death and are frequently accompanied by compensatory processes. Overall, the BM molecules have a crucial role in the careful maintenance of intestinal homeostasis.
Collapse
|
43
|
Abstract
Laminins are cell adhesion molecules that comprise a family of glycoproteins found predominantly in basement membranes, which are the thin sheets of extracellular matrix that underlie epithelial and endothelial cells and surround muscle cells, Schwann cells, and fat cells. Many laminins self-assemble to form networks that remain in close association with cells through interactions with cell surface receptors. Laminins are vital for many physiological functions. They are essential for early embryonic development and organogenesis and have crucial functions in several tissues including muscle, nerve, skin, kidney, lung, and the vasculature. A great wealth of data on laminins is available, and an in-depth description is not attempted here. In this review, I will instead provide a snapshot of laminin structure, tissue distribution, and interactions with other matrix molecules and receptors and briefly describe laminin mutations in mice and humans. Several illuminating and timely reviews are cited that can be consulted for references to original articles and more detailed information concerning laminins.
Collapse
|
44
|
Medeiros RB, Papenfuss KJ, Hoium B, Coley K, Jadrich J, Goh SK, Elayaperumal A, Herrera JE, Resnik E, Ni HT. Novel sequential ChIP and simplified basic ChIP protocols for promoter co-occupancy and target gene identification in human embryonic stem cells. BMC Biotechnol 2009; 9:59. [PMID: 19563662 PMCID: PMC2709612 DOI: 10.1186/1472-6750-9-59] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Accepted: 06/29/2009] [Indexed: 02/08/2023] Open
Abstract
Background The investigation of molecular mechanisms underlying transcriptional regulation, particularly in embryonic stem cells, has received increasing attention and involves the systematic identification of target genes and the analysis of promoter co-occupancy. High-throughput approaches based on chromatin immunoprecipitation (ChIP) have been widely used for this purpose. However, these approaches remain time-consuming, expensive, labor-intensive, involve multiple steps, and require complex statistical analysis. Advances in this field will greatly benefit from the development and use of simple, fast, sensitive and straightforward ChIP assay and analysis methodologies. Results We initially developed a simplified, basic ChIP protocol that combines simplicity, speed and sensitivity. ChIP analysis by real-time PCR was compared to analysis by densitometry with the ImageJ software. This protocol allowed the rapid identification of known target genes for SOX2, NANOG, OCT3/4, SOX17, KLF4, RUNX2, OLIG2, SMAD2/3, BMI-1, and c-MYC in a human embryonic stem cell line. We then developed a novel Sequential ChIP protocol to investigate in vivo promoter co-occupancy, which is basically characterized by the absence of antibody-antigen disruption during the assay. It combines centrifugation of agarose beads and magnetic separation. Using this Sequential ChIP protocol we found that c-MYC associates with the SOX2/NANOG/OCT3/4 complex and identified a novel RUNX2/BMI-1/SMAD2/3 complex in BG01V cells. These two TF complexes associate with two distinct sets of target genes. The RUNX2/BMI-1/SMAD2/3 complex is associated predominantly with genes not expressed in undifferentiated BG01V cells, consistent with the reported role of those TFs as transcriptional repressors. Conclusion These simplified basic ChIP and novel Sequential ChIP protocols were successfully tested with a variety of antibodies with human embryonic stem cells, generated a number of novel observations for future studies and might be useful for high-throughput ChIP-based assays.
Collapse
Affiliation(s)
- Ricardo B Medeiros
- Dept, Antibody Applications and Stem Cells, R&D Systems, Inc,, Minneapolis-MN, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Palasingam P, Jauch R, Ng CKL, Kolatkar PR. The structure of Sox17 bound to DNA reveals a conserved bending topology but selective protein interaction platforms. J Mol Biol 2009; 388:619-30. [PMID: 19328208 DOI: 10.1016/j.jmb.2009.03.055] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 03/17/2009] [Accepted: 03/20/2009] [Indexed: 11/28/2022]
Abstract
Sox17 regulates endodermal lineage commitment and is thought to function antagonistically to the pluripotency determinant Sox2. To investigate the biochemical basis for the distinct functions of Sox2 and Sox17, we solved the crystal structure of the high mobility group domain of Sox17 bound to a DNA element derived from the Lama1 enhancer using crystals diffracting to 2.7 A resolution. Sox17 targets the minor groove and bends the DNA by approximately 80 degrees . The DNA architecture closely resembles the one seen for Sox2/DNA structures, suggesting that the degree of bending is conserved between both proteins and nucleotide substitutions have only marginal effects on the bending topology. Accordingly, affinities of Sox2 and Sox17 for the Lama1 element were found to be identical. However, when the Oct1 contact interface of Sox2 is compared with the corresponding region of Sox17, a significantly altered charge distribution is observed, suggesting differential co-factor recruitment that may explain their biological distinctiveness.
Collapse
Affiliation(s)
- Paaventhan Palasingam
- Laboratory of Structural Biochemistry, Genome Institute of Singapore, 60 Biopolis Street, Singapore, Singapore
| | | | | | | |
Collapse
|
46
|
Qu XB, Pan J, Zhang C, Huang SY. Sox17 facilitates the differentiation of mouse embryonic stem cells into primitive and definitive endoderm in vitro. Dev Growth Differ 2009; 50:585-93. [PMID: 19238729 DOI: 10.1111/j.1440-169x.2008.01056.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The Sox family of HMG (high mobility group)-box transcription factors are highly conserved in vertebrates. Sox members are involved in various developmental processes. Among them Sox17 has been demonstrated to function as an endoderm determinant in zebrafish and Xenopus, respectively. However, little is known about the role of Sox17 in mouse embryonic stem cell (ESC) differentiation. In our research, we investigated the effect of Sox17 on mouse ESC and embryoid body (EB) differentiation. The results demonstrated that Sox17 overexpression upregulated a set of endoderm-specific gene markers, suggesting that Sox17 overexpression induced an ESC differentiation program towards both primitive and definitive endoderm. We believe this finding brings new insights into the understanding of ESC differentiation and the organogenesis of endodermal derivatives.
Collapse
Affiliation(s)
- Xue-Bin Qu
- Key Laboratory of Animal Resistance Research, College of Life Science, Shandong Normal University, 88 East Wenhua Road, Jinan City, Shandong Province, China
| | | | | | | |
Collapse
|
47
|
Navarro-Martín L, Galay-Burgos M, Sweeney G, Piferrer F. Different sox17 transcripts during sex differentiation in sea bass, Dicentrarchus labrax. Mol Cell Endocrinol 2009; 299:240-51. [PMID: 19071190 DOI: 10.1016/j.mce.2008.11.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 11/07/2008] [Accepted: 11/09/2008] [Indexed: 01/11/2023]
Abstract
Sox genes participate in several developmental processes, including sex determination and differentiation. In this study, the genomic structure of sox17 was characterized in the sea bass (sb). Two transcripts, one producing a normal protein (sb Sox17) and another producing a truncated protein (sb t-Sox17) were detected. A third, novel transcript, originated by intron retention (sb i-sox17) was also observed. Sb sox17 was widely distributed, whereas sb i-sox17 was predominantly found in skin and brain. In gonads, sb sox17 expression first increased at 150 days of age, coinciding with the onset of sex differentiation. At 250 days and onwards, sb sox17 expression was significantly higher in females, and mRNA levels correlated with those of gonadal aromatase. Thus, this study provides the first evidence for the presence of alternative splicing by intron retention in a Sox17 gene, and for sex-related differences in expression, implicating sox17 in ovarian development and function in fish.
Collapse
Affiliation(s)
- Laia Navarro-Martín
- Institut de Ciències del Mar, Consejo Superior de Investigaciones Científicas (CSIC), Passeig Marítim, 37-49, 08003 Barcelona, Spain
| | | | | | | |
Collapse
|
48
|
Palmer MB, Majumder P, Cooper JC, Yoon H, Wade PA, Boss JM. Yin yang 1 regulates the expression of snail through a distal enhancer. Mol Cancer Res 2009; 7:221-9. [PMID: 19208738 DOI: 10.1158/1541-7786.mcr-08-0229] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Expression of the Snail gene is required for the epithelial-mesenchymal transitions that accompany mammalian gastrulation, neural crest migration, and organ formation. Pathologic expression of Snail contributes to the migratory capacity of invasive tumors, including melanomas. To investigate the mechanism of Snail up-regulation in human melanoma cells, a conserved enhancer located 3' of the Snail gene was analyzed. An overlapping Ets and yin yang 1 (YY1) consensus sequence, in addition to a SOX consensus sequence, was required for full enhancer activity. Proteins specifically binding these sequences were detected by electrophoretic mobility shift assay. The Ets/YY1 binding activity was purified by DNA-affinity chromatography and identified as YY1. Although ubiquitously expressed, YY1 was bound at the Snail 3' enhancer in vivo in Snail-expressing cells but not in cells that did not express Snail. Knockdown of YY1 in A375 cells led to decreased Snail expression. These results identify a role for YY1 in regulating transcription of Snail in melanoma cells through binding to the Snail 3' enhancer.
Collapse
Affiliation(s)
- Matthew B Palmer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
49
|
Martinez-Ceballos E, Gudas LJ. Hoxa1 is required for the retinoic acid-induced differentiation of embryonic stem cells into neurons. J Neurosci Res 2008; 86:2809-19. [PMID: 18512762 DOI: 10.1002/jnr.21729] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ability of embryonic stem (ES) cells to differentiate into different cell fates has been extensively evaluated, and several protocols exist for the generation of various types of cells from mouse and human ES cells. We used a differentiation protocol that involves embryoid body formation and all-trans-retinoic acid (RA, 5 microM) treatment (EB/5 microM RA) to test the ability of Hoxa1 null ES cells to adopt a neuronal fate. Hoxa1(-/-) ES cells, when treated in this EB/5 microM RA protocol, failed to differentiate along a neural lineage; Hoxa1(-/-) ES cells express severalfold lower levels of many neuronal differentiation markers, including nestin, beta-tubulin III, and MAP2, and conversely, higher levels of endodermal differentiation markers (i.e., Sox17, Col4a1) than wild type (Wt) cells. Reintroduction of exogenous Hoxa1, under the control of the metallothionein I promoter, into Hoxa1(-/-) ES cells restored their capacity to generate neurons. Moreover, overexpression of Sox17, a gene that regulates endodermal differentiation, in Wt ES cells resulted in endodermal differentiation and in a complete abolition of beta-tubulin III expression. Thus, Hoxa1 activity is essential for the neuronal differentiation of ES cells in the presence of all-trans-RA, and Hoxa1 may promote neural differentiation by inhibiting Sox17 expression. Pharmacological manipulation of Hoxa1 levels may provide a method for promoting neuronal differentiation for therapeutic uses. Furthermore, because mutations in the Hoxa1 gene can cause autism spectrum disorder in humans, these data also provide important mechanistic insights into the early developmental processes that may result in this disorder.
Collapse
|
50
|
Ng CKL, Palasingam P, Venkatachalam R, Baburajendran N, Cheng J, Jauch R, Kolatkar PR. Purification, crystallization and preliminary X-ray diffraction analysis of the HMG domain of Sox17 in complex with DNA. Acta Crystallogr Sect F Struct Biol Cryst Commun 2008; 64:1184-7. [PMID: 19052383 PMCID: PMC2593696 DOI: 10.1107/s1744309108038724] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Accepted: 11/19/2008] [Indexed: 11/10/2022]
Abstract
Sox17 is a member of the SRY-related high-mobility group (HMG) of transcription factors that have been shown to direct endodermal differentiation in early mammalian development. The LAMA1 gene encoding the alpha-chain of laminin-1 has been reported to be directly bound and regulated by Sox17. This paper describes the details of initial crystallization attempts with the HMG domain of mouse Sox17 (mSox17-HMG) with a 16-mer DNA element derived from the LAMA1 enhancer and optimization strategies to obtain a better diffracting crystal. The best diffracting crystal was obtained in a condition containing 0.1 M Tris-HCl pH 7.4, 0.2 M MgCl(2), 30% PEG 3350 using the hanging-drop vapour-diffusion method. A highly redundant in-house data set was collected to 2.75 A resolution with 99% completeness. The presence of the mSox17-HMG-DNA complex within the crystals was confirmed and Matthews analysis indicated the presence of one complex per asymmetric unit.
Collapse
Affiliation(s)
- Calista Keow Leng Ng
- Laboratory for Structural Biochemistry, Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore 138672, Singapore
| | - Paaventhan Palasingam
- Laboratory for Structural Biochemistry, Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore 138672, Singapore
| | - Rajakannan Venkatachalam
- Department of Structural Biology, Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Nithya Baburajendran
- Laboratory for Structural Biochemistry, Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore 138672, Singapore
| | - Jason Cheng
- School of Information Technology and Applied Sciences, Temasek Polytechnic, 21 Tampines Avenue 1, Singapore 529757, Singapore
| | - Ralf Jauch
- Laboratory for Structural Biochemistry, Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore 138672, Singapore
| | - Prasanna R. Kolatkar
- Laboratory for Structural Biochemistry, Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore 138672, Singapore
| |
Collapse
|