1
|
Meharwade T, Joumier L, Parisotto M, Huynh V, Lummertz da Rocha E, Malleshaiah M. Cross-activation of FGF, NODAL, and WNT pathways constrains BMP-signaling-mediated induction of the totipotent state in mouse embryonic stem cells. Cell Rep 2023; 42:112438. [PMID: 37126449 DOI: 10.1016/j.celrep.2023.112438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 11/11/2022] [Accepted: 04/11/2023] [Indexed: 05/02/2023] Open
Abstract
Embryonic stem cells (ESCs) are an attractive model to study the relationship between signaling and cell fates. Cultured mouse ESCs can exist in multiple states resembling distinct stages of early embryogenesis, such as totipotent, pluripotent, primed, and primitive endoderm. The signaling mechanisms regulating the totipotent state and coexistence of these states are poorly understood. Here we identify bone morphogenetic protein (BMP) signaling as an inducer of the totipotent state. However, we discover that BMP's role is constrained by the cross-activation of FGF, NODAL, and WNT pathways. We exploit this finding to enhance the proportion of totipotent cells by rationally inhibiting the cross-activated pathways. Single-cell mRNA sequencing reveals that induction of the totipotent state is accompanied by suppression of primed and primitive endoderm states. Furthermore, reprogrammed totipotent cells we generate in culture resemble totipotent cells of preimplantation embryo. Our findings reveal a BMP signaling mechanism regulating both the totipotent state and heterogeneity of ESCs.
Collapse
Affiliation(s)
- Thulaj Meharwade
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, C.P. 6128, Succursale Centre-ville, Montreal, QC H3C 3J7, Canada
| | - Loïck Joumier
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, C.P. 6128, Succursale Centre-ville, Montreal, QC H3C 3J7, Canada
| | - Maxime Parisotto
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
| | - Vivian Huynh
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada; Molecular Biology Program, University of Montreal, C.P. 6128, Succursale Centre-ville, Montreal, QC H3C 3J7, Canada
| | - Edroaldo Lummertz da Rocha
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Mohan Malleshaiah
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, C.P. 6128, Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; Molecular Biology Program, University of Montreal, C.P. 6128, Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; The Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; McGill Regenerative Medicine Network, 1160 Pine Avenue West, Montreal, QC H3A 1A3, Canada.
| |
Collapse
|
2
|
Gao F, Wu S, Li Y, Fang Y, Liu M, Du J, Kong Q, An T. Inhibition of TGF-β pathway improved the pluripotency of porcine pluripotent stem cells. In Vitro Cell Dev Biol Anim 2023; 59:142-152. [PMID: 36867291 DOI: 10.1007/s11626-023-00752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/09/2023] [Indexed: 03/04/2023]
Abstract
Porcine pluripotent stem cells had been derived from different culture systems. PeNK6 is a porcine pluripotent stem cell line that we established from an E5.5 embryo in a defined culture system. Signaling pathways related with pluripotency had been assessed in this cell line, and TGF-β signaling pathway-related genes were found upregulated significantly. In this study, we elucidated the role of the TGF-β signaling pathway in PeNK6 through adding small molecule inhibitors, SB431542 (KOSB) or A83-01 (KOA), into the original culture medium (KO) and analyzing the expression and activity of key factors involved in the TGF-β signaling pathway. In KOSB/KOA medium, the morphology of PeNK6 became compact and the nuclear-to-cytoplasm ratio was increased. The expression of the core transcription factor SOX2 was significantly upregulated compared with cell lines in the control KO medium, and the differentiation potential became balanced among three germ layers rather than bias to neuroectoderm/endoderm as the original PeNK6 did. The results indicated that inhibition of TGF-β has positive effects on the porcine pluripotency. Based on these results, we established a pluripotent cell line (PeWKSB) from E5.5 blastocyst by employing TGF-β inhibitors, and the cell line showed improved pluripotency.
Collapse
Affiliation(s)
- Fang Gao
- College of Life Science, Northeast Forestry University, Harbin, 150040, People's Republic of China.,Laboratory of Embryo Biotechnology, College of Life Science, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shuang Wu
- Laboratory of Embryo Biotechnology, College of Life Science, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Yan Li
- Laboratory of Embryo Biotechnology, College of Life Science, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Yuan Fang
- Laboratory of Embryo Biotechnology, College of Life Science, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Minli Liu
- Laboratory of Embryo Biotechnology, College of Life Science, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Jiawei Du
- Laboratory of Embryo Biotechnology, College of Life Science, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Qingran Kong
- Laboratory of Embryo Biotechnology, College of Life Science, Northeast Agricultural University, Harbin, 150030, People's Republic of China.,Wenzhou Medical University, Wenzhou, 325035, People's Republic of China
| | - Tiezhu An
- College of Life Science, Northeast Forestry University, Harbin, 150040, People's Republic of China.
| |
Collapse
|
3
|
Secco B, Saitoski K, Drareni K, Soprani A, Pechberty S, Rachdi L, Venteclef N, Scharfmann R. Loss of Human Beta Cell Identity in a Reconstructed Omental Stromal Cell Environment. Cells 2022; 11:cells11060924. [PMID: 35326375 PMCID: PMC8946101 DOI: 10.3390/cells11060924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/30/2022] Open
Abstract
In human type 2 diabetes, adipose tissue plays an important role in disturbing glucose homeostasis by secreting factors that affect the function of cells and tissues throughout the body, including insulin-producing pancreatic beta cells. We aimed here at studying the paracrine effect of stromal cells isolated from subcutaneous and omental adipose tissue on human beta cells. We developed an in vitro model wherein the functional human beta cell line EndoC-βH1 was treated with conditioned media from human adipose tissues. By using RNA-sequencing and western blotting, we determined that a conditioned medium derived from omental stromal cells stimulates several pathways, such as STAT, SMAD and RELA, in EndoC-βH1 cells. We also observed that upon treatment, the expression of beta cell markers decreased while dedifferentiation markers increased. Loss-of-function experiments that efficiently blocked specific signaling pathways did not reverse dedifferentiation, suggesting the implication of more than one pathway in this regulatory process. Taken together, we demonstrate that soluble factors derived from stromal cells isolated from human omental adipose tissue signal human beta cells and modulate their identity.
Collapse
Affiliation(s)
- Blandine Secco
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Kevin Saitoski
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Karima Drareni
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Université de Paris, 75006 Paris, France; (K.D.); (A.S.); (N.V.)
| | - Antoine Soprani
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Université de Paris, 75006 Paris, France; (K.D.); (A.S.); (N.V.)
- Clinique Geoffroy Saint-Hilaire, Ramsey General de Santé, 75005 Paris, France
| | - Severine Pechberty
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Latif Rachdi
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Nicolas Venteclef
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Université de Paris, 75006 Paris, France; (K.D.); (A.S.); (N.V.)
| | - Raphaël Scharfmann
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
- Correspondence: ; Tel.: +(33)-1-76-53-55-68
| |
Collapse
|
4
|
Mayère C, Neirijnck Y, Sararols P, Rands CM, Stévant I, Kühne F, Chassot AA, Chaboissier MC, Dermitzakis ET, Nef S. Single-cell transcriptomics reveal temporal dynamics of critical regulators of germ cell fate during mouse sex determination. FASEB J 2021; 35:e21452. [PMID: 33749946 DOI: 10.1096/fj.202002420r] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/22/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022]
Abstract
Despite the importance of germ cell (GC) differentiation for sexual reproduction, the gene networks underlying their fate remain unclear. Here, we comprehensively characterize the gene expression dynamics during sex determination based on single-cell RNA sequencing of 14 914 XX and XY mouse GCs between embryonic days (E) 9.0 and 16.5. We found that XX and XY GCs diverge transcriptionally as early as E11.5 with upregulation of genes downstream of the bone morphogenic protein (BMP) and nodal/Activin pathways in XY and XX GCs, respectively. We also identified a sex-specific upregulation of genes associated with negative regulation of mRNA processing and an increase in intron retention consistent with a reduction in mRNA splicing in XY testicular GCs by E13.5. Using computational gene regulation network inference analysis, we identified sex-specific, sequential waves of putative key regulator genes during GC differentiation and revealed that the meiotic genes are regulated by positive and negative master modules acting in an antagonistic fashion. Finally, we found that rare adrenal GCs enter meiosis similarly to ovarian GCs but display altered expression of master genes controlling the female and male genetic programs, indicating that the somatic environment is important for GC function. Our data are available on a web platform and provide a molecular roadmap of GC sex determination at single-cell resolution, which will serve as a valuable resource for future studies of gonad development, function, and disease.
Collapse
Affiliation(s)
- Chloé Mayère
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.,iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Yasmine Neirijnck
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.,CNRS, Inserm, iBV, Université Côte d'Azur, Nice, France
| | - Pauline Sararols
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Chris M Rands
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Isabelle Stévant
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.,iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Françoise Kühne
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | | | | | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.,iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.,iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| |
Collapse
|
5
|
Transient Nodal Signaling in Left Precursors Coordinates Opposed Asymmetries Shaping the Heart Loop. Dev Cell 2020; 55:413-431.e6. [PMID: 33171097 DOI: 10.1016/j.devcel.2020.10.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 07/17/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
The secreted factor Nodal, known as a major left determinant, is associated with severe heart defects. Yet, it has been unclear how it regulates asymmetric morphogenesis such as heart looping, which align cardiac chambers to establish the double blood circulation. Here, we report that Nodal is transiently active in precursors of the mouse heart tube poles, before looping. In conditional mutants, we show that Nodal is not required to initiate asymmetric morphogenesis. We provide evidence of a heart-specific random generator of asymmetry that is independent of Nodal. Using 3D quantifications and simulations, we demonstrate that Nodal functions as a bias of this mechanism: it is required to amplify and coordinate opposed left-right asymmetries at the heart tube poles, thus generating a robust helical shape. We identify downstream effectors of Nodal signaling, regulating asymmetries in cell proliferation, differentiation, and extracellular matrix composition. Our study uncovers how Nodal regulates asymmetric organogenesis.
Collapse
|
6
|
Moazzeni H, Khani M, Elahi E. Insights into the regulatory molecules involved in glaucoma pathogenesis. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:782-827. [PMID: 32935930 DOI: 10.1002/ajmg.c.31833] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022]
Abstract
Glaucoma is an important cause of irreversible blindness, characterized by optic nerve anomalies. Increased intraocular pressure (IOP) and aging are major risk factors. Retinal ganglion cells and trabecular meshwork cells are certainly involved in the etiology of glaucoma. Glaucoma is usually a complex disease, and various genes and functions may contribute to its etiology. Among these may be genes that encode regulatory molecules. In this review, regulatory molecules including 18 transcription factors (TFs), 195 microRNAs (miRNAs), 106 long noncoding RNAs (lncRNAs), and two circular RNAs (circRNAs) that are reasonable candidates for having roles in glaucoma pathogenesis are described. The targets of the regulators are reported. Glaucoma-related features including apoptosis, stress responses, immune functions, ECM properties, IOP, and eye development are affected by the targeted genes. The targeted genes that are frequently targeted by multiple regulators most often affect apoptosis and the related features of cell death and cell survival. BCL2, CDKN1A, and TP53 are among the frequent targets of three types of glaucoma-relevant regulators, TFs, miRNAs, and lncRNAs. TP53 was itself identified as a glaucoma-relevant TF. Several of the glaucoma-relevant TFs are themselves among frequent targets of regulatory molecules, which is consistent with existence of a complex network involved in glaucoma pathogenesis.
Collapse
Affiliation(s)
- Hamidreza Moazzeni
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Marzieh Khani
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
7
|
Moazzeni H, Mirrahimi M, Moghadam A, Banaei-Esfahani A, Yazdani S, Elahi E. Identification of genes involved in glaucoma pathogenesis using combined network analysis and empirical studies. Hum Mol Genet 2019; 28:3637-3663. [PMID: 31518395 DOI: 10.1093/hmg/ddz222] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/30/2019] [Accepted: 09/04/2019] [Indexed: 12/25/2022] Open
Abstract
Glaucoma is a leading cause of blindness. We aimed in this study to identify genes that may make subtle and cumulative contributions to glaucoma pathogenesis. To this end, we identified molecular interactions and pathways that include transcription factors (TFs) FOXC1, PITX2, PAX6 and NFKB1 and various microRNAs including miR-204 known to have relevance to trabecular meshwork (TM) functions and/or glaucoma. TM tissue is involved in glaucoma pathogenesis. In-house microarray transcriptome results and data sources were used to identify target genes of the regulatory molecules. Bioinformatics analyses were done to filter TM and glaucoma relevant genes. These were submitted to network-creating softwares to define interactions, pathways and a network that would include the genes. The network was stringently scrutinized and minimized, then expanded by addition of microarray data and data on TF and microRNA-binding sites. Selected features of the network were confirmed by empirical studies such as dual luciferase assays, real-time PCR and western blot experiments and apoptosis assays. MYOC, WDR36, LTPBP2, RHOA, CYP1B1, OPA1, SPARC, MEIS2, PLEKHG5, RGS5, BBS5, ALDH1A1, NOMO2, CXCL6, FMNL2, ADAMTS5, CLOCK and DKK1 were among the genes included in the final network. Pathways identified included those that affect ECM properties, IOP, ciliary body functions, retinal ganglion cell viability, apoptosis, focal adhesion and oxidative stress response. The identification of many genes potentially involved in glaucoma pathology is consistent with its being a complex disease. The inclusion of several known glaucoma-related genes validates the approach used.
Collapse
Affiliation(s)
- Hamidreza Moazzeni
- School of Biology, College of Science, University of Tehran, Tehran, Iran
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehraban Mirrahimi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Abolfazl Moghadam
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Amir Banaei-Esfahani
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Shahin Yazdani
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
8
|
Li Y, Luo W, Yang W. Nuclear Transport and Accumulation of Smad Proteins Studied by Single-Molecule Microscopy. Biophys J 2019; 114:2243-2251. [PMID: 29742417 DOI: 10.1016/j.bpj.2018.03.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 03/15/2018] [Indexed: 12/24/2022] Open
Abstract
Nuclear translocation of stimulated Smad heterocomplexes is a critical step in the signal transduction of transforming growth factor β (TGF-β) from transmembrane receptors into the nucleus. Specifically, normal nuclear accumulation of Smad2/Smad4 heterocomplexes induced by TGF-β1 is involved in carcinogenesis. However, the relationship between nuclear accumulation and the nucleocytoplasmic transport kinetics of Smad proteins in the presence of TGF-β1 remains obscure. By combining a high-speed single-molecule tracking microscopy and Förster resonance energy transfer technique, we tracked the entire TGF-β1-induced process of Smad2/Smad4 heterocomplex formation, as well as their transport through nuclear pore complexes in live cells, with a high single-molecule localization precision of 2 ms and <20 nm. Our single-molecule Förster resonance energy transfer data have revealed that in TGF-β1-treated cells, Smad2/Smad4 heterocomplexes formed in the cytoplasm, imported through the nuclear pore complexes as entireties, and finally dissociated in the nucleus. Moreover, we found that basal-state Smad2 or Smad4 cannot accumulate in the nucleus without the presence of TGF-β1, mainly because both of them have an approximately twofold higher nuclear export efficiency compared to their nuclear import. Remarkably and reversely, heterocomplexes of Smad2/Smad4 induced by TGF-β1 can rapidly concentrate in the nucleus because of their almost fourfold higher nuclear import rate in comparison with their nuclear export rate. Thus, we believe that the determined TGF-β1-dependent transport configurations and efficiencies for the basal-state Smad or stimulated Smad heterocomplexes elucidate the basic molecular mechanism to understand their nuclear transport and accumulation.
Collapse
Affiliation(s)
- Yichen Li
- Department of Biology, Temple University, Philadelphia, Pennsylvania
| | - Wangxi Luo
- Department of Biology, Temple University, Philadelphia, Pennsylvania
| | - Weidong Yang
- Department of Biology, Temple University, Philadelphia, Pennsylvania.
| |
Collapse
|
9
|
Xu Y, Luo X, Fang Z, Zheng X, Zeng Y, Zhu C, Gu J, Tang F, Hu Y, Hu G, Jin Y, Li H. Transcription coactivator Cited1 acts as an inducer of trophoblast-like state from mouse embryonic stem cells through the activation of BMP signaling. Cell Death Dis 2018; 9:924. [PMID: 30206204 PMCID: PMC6134011 DOI: 10.1038/s41419-018-0991-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 07/16/2018] [Accepted: 08/01/2018] [Indexed: 12/31/2022]
Abstract
Trophoblast lineages, precursors of the placenta, are essential for post-implantation embryo survival. However, the regulatory network of trophoblast development remains incompletely understood. Here, we report that Cited1, a transcription coactivator, is a robust inducer for trophoblast-like state from mouse embryonic stem cells (ESCs). Depletion of Cited1 in ESCs compromises the trophoblast lineage specification induced by BMP signaling. In contrast, overexpression of Cited1 in ESCs induces a trophoblast-like state with elevated expression of trophoblast marker genes in vitro and generation of trophoblastic tumors in vivo. Furthermore, global transcriptome profile analysis indicates that ectopic Cited1 activates a trophoblast-like transcriptional program in ESCs. Mechanistically, Cited1 interacts with Bmpr2 and Smad4 to activate the Cited1–Bmpr2–Smad1/5/8 axis in the cytoplasm and Cited1–Smad4–p300 complexes in the nucleus, respectively. Collectively, our results show that Cited1 plays an important role in regulating trophoblast lineage specification through activating the BMP signaling pathway.
Collapse
Affiliation(s)
- Yanli Xu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Xinlong Luo
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China.,KU Leuven Department of Development and Regeneration, Stem Cell Institute Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Zhuoqing Fang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, 200032, Shanghai, China
| | - Xiaofeng Zheng
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Yanwu Zeng
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Chaonan Zhu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Junjie Gu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Fan Tang
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Yanqin Hu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Ying Jin
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China. .,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, 200032, Shanghai, China.
| | - Hui Li
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China.
| |
Collapse
|
10
|
Bragança J. SMAD2/3, versatile molecular tools for cellular engineering. Stem Cell Investig 2018; 5:24. [PMID: 30148157 DOI: 10.21037/sci.2018.07.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 07/17/2018] [Indexed: 01/01/2023]
Affiliation(s)
- José Bragança
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal.,Centre for Biomedical Research-CBMR, University of Algarve, Campus of Gambelas, 8005-139 Faro, Portugal.,ABC - Algarve Biomedical Centre, 8005-139 Faro, Portugal
| |
Collapse
|
11
|
Abstract
TGF-β family ligands function in inducing and patterning many tissues of the early vertebrate embryonic body plan. Nodal signaling is essential for the specification of mesendodermal tissues and the concurrent cellular movements of gastrulation. Bone morphogenetic protein (BMP) signaling patterns tissues along the dorsal-ventral axis and simultaneously directs the cell movements of convergence and extension. After gastrulation, a second wave of Nodal signaling breaks the symmetry between the left and right sides of the embryo. During these processes, elaborate regulatory feedback between TGF-β ligands and their antagonists direct the proper specification and patterning of embryonic tissues. In this review, we summarize the current knowledge of the function and regulation of TGF-β family signaling in these processes. Although we cover principles that are involved in the development of all vertebrate embryos, we focus specifically on three popular model organisms: the mouse Mus musculus, the African clawed frog of the genus Xenopus, and the zebrafish Danio rerio, highlighting the similarities and differences between these species.
Collapse
Affiliation(s)
- Joseph Zinski
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Benjamin Tajer
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Mary C Mullins
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| |
Collapse
|
12
|
Zhang N, Lyu Y, Pan X, Xu L, Xuan A, He X, Huang W, Long D. miR‑146b‑5p promotes the neural conversion of pluripotent stem cells by targeting Smad4. Int J Mol Med 2017; 40:814-824. [PMID: 28713933 PMCID: PMC5548013 DOI: 10.3892/ijmm.2017.3064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/30/2017] [Indexed: 01/06/2023] Open
Abstract
Pluripotent stem cells (PSCs) are regarded as potential sources that provide specific neural cells for cell therapy in some nervous system diseases. However, the mechanisms underlying the neural differentiation of PSCs remain largely unknown. MicroRNAs (miRNAs or miRs) are a class of small non-protein-coding RNAs that act as critical regulatory molecules in many cellular processes. In this study, we found that miR-146b-5p expression was markedly increased following the neural induction of mouse embryonic stem cells (ESCs) or induced PSCs (iPSCs). In this study, to further identify the role of miR-146b-5p, we generated stable miR-146b-5p- overexpressing ESC and iPSC cell lines, and induced the differentiation of these cells by the adherent monolayer culture method. In the miR-146b-5p-overexpressing ESC- or iPSC- derived cultures, RT-qPCR analysis revealed that the mRNA expression levels of neuroectoderm markers, such as Sox1, Nestin and Pax6, were markedly increased, and flow cytometric analysis verified that the number of Nestin-positive cells was higher in the miR-146b-5p-overexpressing compared with the control cells. Mechanistically, the miR-146b-5p-overexpressing ESCs or iPSCs exhibited a significant reduction in Oct4 expression, which may be an explanation for these cells having a tendency to differentiate towards the neural lineage. Moreover, we confirmed that miR-146b-5p directly targeted Smad4 and negatively regulated the transforming growth factor (TGF)-β signaling pathway, which contributed to the neural commitment of PSCs. Collectively, our findings uncover the essential role of miR-146b-5p in the neural conversion of PSCs.
Collapse
Affiliation(s)
- Nianping Zhang
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Ying Lyu
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xuebing Pan
- Department of Human Anatomy, College of Health Sciences of Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Liping Xu
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Aiguo Xuan
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xiaosong He
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Wandan Huang
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Dahong Long
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| |
Collapse
|
13
|
Menchero S, Rayon T, Andreu MJ, Manzanares M. Signaling pathways in mammalian preimplantation development: Linking cellular phenotypes to lineage decisions. Dev Dyn 2016; 246:245-261. [DOI: 10.1002/dvdy.24471] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/16/2016] [Accepted: 11/16/2016] [Indexed: 12/20/2022] Open
Affiliation(s)
- Sergio Menchero
- Centro Nacional de Investigaciones Cardiovasculares (CNIC); Madrid Spain
| | - Teresa Rayon
- Centro Nacional de Investigaciones Cardiovasculares (CNIC); Madrid Spain
| | - Maria Jose Andreu
- Centro Nacional de Investigaciones Cardiovasculares (CNIC); Madrid Spain
| | - Miguel Manzanares
- Centro Nacional de Investigaciones Cardiovasculares (CNIC); Madrid Spain
| |
Collapse
|
14
|
Russell R, Perkhofer L, Liebau S, Lin Q, Lechel A, Feld FM, Hessmann E, Gaedcke J, Güthle M, Zenke M, Hartmann D, von Figura G, Weissinger SE, Rudolph KL, Möller P, Lennerz JK, Seufferlein T, Wagner M, Kleger A. Loss of ATM accelerates pancreatic cancer formation and epithelial-mesenchymal transition. Nat Commun 2015; 6:7677. [PMID: 26220524 PMCID: PMC4532798 DOI: 10.1038/ncomms8677] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 05/30/2015] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with accumulation of particular oncogenic mutations and recent genetic sequencing studies have identified ataxia telangiectasia-mutated (ATM) mutations in PDAC cohorts. Here we report that conditional deletion of ATM in a mouse model of PDAC induces a greater number of proliferative precursor lesions coupled with a pronounced fibrotic reaction. ATM-targeted mice display altered TGFβ-superfamily signalling and enhanced epithelial-to-mesenchymal transition (EMT) coupled with shortened survival. Notably, our mouse model recapitulates many features of more aggressive human PDAC subtypes. Particularly, we report that low expression of ATM predicts EMT, a gene signature specific for Bmp4 signalling and poor prognosis in human PDAC. Our data suggest an intimate link between ATM expression and pancreatic cancer progression in mice and men.
Collapse
Affiliation(s)
- Ronan Russell
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Lukas Perkhofer
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Stefan Liebau
- Institute of Neuroanatomy, Eberhard Karls University Tuebingen, Oesterbergstr. 3, Tuebingen 72074, Germany
| | - Qiong Lin
- Department of Cell Biology, Institute for Biomedical Engineering, Medical Faculty, RWTH Aachen University, Pauwelstr. 30, Aachen 52074, Germany
| | - André Lechel
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Fenja M Feld
- Institute of Pathology, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Elisabeth Hessmann
- Department of Gastroenterology II, University Medical Center Goettingen, Robert-Koch-Str. 40, Goettingen 37075, Germany
| | - Jochen Gaedcke
- Department of Surgery, University Medical Center Goettingen, Robert-Koch-Str. 40, Goettingen 37075, Germany
| | - Melanie Güthle
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Martin Zenke
- Department of Cell Biology, Institute for Biomedical Engineering, Medical Faculty, RWTH Aachen University, Pauwelstr. 30, Aachen 52074, Germany
| | - Daniel Hartmann
- Department of Surgery, Technische Universität München, Ismaninger Str. 22, Munich 81675, Germany
| | - Guido von Figura
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, Munich 81675, Germany
| | | | - Karl-Lenhard Rudolph
- Leibniz Institute for Age Research - Fritz Lipmann Institute e.V., Beutenbergstr. 11, Jena 07745, Germany
| | - Peter Möller
- Institute of Pathology, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Jochen K Lennerz
- Institute of Pathology, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Martin Wagner
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, Ulm 89081, Germany
| |
Collapse
|
15
|
Dubrulle J, Jordan BM, Akhmetova L, Farrell JA, Kim SH, Solnica-Krezel L, Schier AF. Response to Nodal morphogen gradient is determined by the kinetics of target gene induction. eLife 2015; 4. [PMID: 25869585 PMCID: PMC4395910 DOI: 10.7554/elife.05042] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 03/02/2015] [Indexed: 12/24/2022] Open
Abstract
Morphogen gradients expose cells to different signal concentrations and induce target genes with different ranges of expression. To determine how the Nodal morphogen gradient induces distinct gene expression patterns during zebrafish embryogenesis, we measured the activation dynamics of the signal transducer Smad2 and the expression kinetics of long- and short-range target genes. We found that threshold models based on ligand concentration are insufficient to predict the response of target genes. Instead, morphogen interpretation is shaped by the kinetics of target gene induction: the higher the rate of transcription and the earlier the onset of induction, the greater the spatial range of expression. Thus, the timing and magnitude of target gene expression can be used to modulate the range of expression and diversify the response to morphogen gradients. DOI:http://dx.doi.org/10.7554/eLife.05042.001 How a cell can tell where it is in a developing embryo has fascinated scientists for decades. The pioneering computer scientist and mathematical biologist Alan Turing was the first person to coin the term ‘morphogen’ to describe a protein that provides information about locations in the body. A morphogen is released from a group of cells (called the ‘source’) and as it moves away its activity (called the ‘signal’) declines gradually. Cells sense this signal gradient and use it to detect their position with respect to the source. Nodal is an important morphogen and is required to establish the correct identity of cells in the embryo; for example, it helps determine which cells should become a brain or heart or gut cell and so on. The zebrafish is a widely used model to study animal development, in part because its embryos are transparent; this allows cells and proteins to be easily observed under a microscope. When Nodal acts on cells, another protein called Smad2 becomes activated, moves into the cell's nucleus, and then binds to specific genes. This triggers the expression of these genes, which are first copied into mRNA molecules via a process known as transcription and are then translated into proteins. The protein products of these targeted genes control cell identity and movement. Several models have been proposed to explain how different concentrations of Nodal switch on the expression of different target genes; that is to say, to explain how a cell interprets the Nodal gradient. Dubrulle et al. have now measured factors that underlie how this gradient is interpreted. Individual cells in zebrafish embryos were tracked under a microscope, and Smad2 activation and gene expression were assessed. Dubrulle et al. found that, in contradiction to previous models, the amount of Nodal present on its own was insufficient to predict the target gene response. Instead, their analysis suggests that the size of each target gene's response depends on its rate of transcription and how quickly it is first expressed in response to Nodal. These findings of Dubrulle et al. suggest that timing and transcription rate are important in determining the appropriate response to Nodal. Further work will be now needed to find out whether similar mechanisms regulate other processes that rely on the activity of morphogens. DOI:http://dx.doi.org/10.7554/eLife.05042.002
Collapse
Affiliation(s)
- Julien Dubrulle
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Benjamin M Jordan
- Department of Mathematics, College of Science and Engineering, University of Minnesota, Minneapolis, United States
| | - Laila Akhmetova
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Jeffrey A Farrell
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Seok-Hyung Kim
- Division of Medicine, Medical University of South Carolina, Charleston, United States
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, United States
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| |
Collapse
|
16
|
Gaarenstroom T, Hill CS. TGF-β signaling to chromatin: how Smads regulate transcription during self-renewal and differentiation. Semin Cell Dev Biol 2014; 32:107-18. [PMID: 24503509 DOI: 10.1016/j.semcdb.2014.01.009] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 01/29/2014] [Indexed: 12/20/2022]
Abstract
Ligands of the TGF-β superfamily (including the TGF-βs, Nodal and BMPs) play instructive roles during embryonic development. This is achieved by regulation of genes important for both maintaining pluripotency and germ layer specification and differentiation. Here we review how the TGF-β superfamily ligands signal to the chromatin to regulate transcription during development. The effectors of the pathway, the Smad transcription factors, are regulated in a combinatorial and spatiotemporal manner. This occurs via post-translational modifications affecting stability, localization and activity, as well as through interactions with other transcription factors and chromatin modifying enzymes, which occur on DNA. Expression profiling and Chromatin Immunoprecipitation have defined Smad target genes and binding sites on a genome-wide scale, which vary between cell types and differentiation stages. This has led to the insight that Smad-mediated transcriptional responses are influenced by the presence of master transcription factors, such as OCT4, SOX2 and NANOG in embryonic stem cells, interaction with other signal-induced factors, as well as by the general chromatin remodeling machinery. Interplay with transcriptional repressors and the polycomb group proteins also regulates the balance between expression of self-renewal and mesendoderm-specific genes in embryonic stem cells and during early development.
Collapse
Affiliation(s)
- Tessa Gaarenstroom
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, United Kingdom
| | - Caroline S Hill
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, United Kingdom.
| |
Collapse
|
17
|
Papanayotou C, Benhaddou A, Camus A, Perea-Gomez A, Jouneau A, Mezger V, Langa F, Ott S, Sabéran-Djoneidi D, Collignon J. A novel nodal enhancer dependent on pluripotency factors and smad2/3 signaling conditions a regulatory switch during epiblast maturation. PLoS Biol 2014; 12:e1001890. [PMID: 24960041 PMCID: PMC4068991 DOI: 10.1371/journal.pbio.1001890] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 05/15/2014] [Indexed: 02/07/2023] Open
Abstract
HBE, a newly discovered enhancer element, mediates the influence of pluripotency factors and Activin/Nodal signaling on early Nodal expression in the mouse embryo, and controls the activation of later-acting Nodal enhancers. During early development, modulations in the expression of Nodal, a TGFβ family member, determine the specification of embryonic and extra-embryonic cell identities. Nodal has been extensively studied in the mouse, but aspects of its early expression remain unaccounted for. We identified a conserved hotspot for the binding of pluripotency factors at the Nodal locus and called this sequence “highly bound element” (HBE). Luciferase-based assays, the analysis of fluorescent HBE reporter transgenes, and a conditional mutation of HBE allowed us to establish that HBE behaves as an enhancer, is activated ahead of other Nodal enhancers in the epiblast, and is essential to Nodal expression in embryonic stem cells (ESCs) and in the mouse embryo. We also showed that HBE enhancer activity is critically dependent on its interaction with the pluripotency factor Oct4 and on Activin/Nodal signaling. Use of an in vitro model of epiblast maturation, relying on the differentiation of ESCs into epiblast stem cells (EpiSCs), revealed that this process entails a shift in the regulation of Nodal expression from an HBE-driven phase to an ASE-driven phase, ASE being another autoregulatory Nodal enhancer. Deletion of HBE in ESCs or in EpiSCs allowed us to show that HBE, although not necessary for Nodal expression in EpiSCs, is required in differentiating ESCs to activate the differentiation-promoting ASE and therefore controls this regulatory shift. Our findings clarify how early Nodal expression is regulated and suggest how this regulation can promote the specification of extra-embryonic precusors without inducing premature differentiation of epiblast cells. More generally, they open new perspectives on how pluripotency factors achieve their function. In the early mouse embryo, Nodal, a member of the TGFbeta superfamily of signalling proteins, promotes the differentiation of extra-embryonic tissues, as well as tissues within the developing embryo itself. Characterising the regulation of Nodal gene expression is essential to understand how Nodal signals in diverse tissue types and at different stages of embryonic development. Four distinct enhancer sequences have been shown to regulate Nodal expression, although none could account for it in the preimplantation epiblast or in embryonic stem cells. We identified a novel enhancer, HBE, responsible for the earliest aspects of Nodal expression. We show that activation of HBE depends on its interaction with a well-known pluripotency factor called Oct4. HBE itself also controls the activation of at least one other Nodal enhancer. Our findings clarify how early Nodal expression is regulated and reveal how pluripotency factors may control the onset of differentiation in embryonic tissues.
Collapse
Affiliation(s)
- Costis Papanayotou
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
- * E-mail: (JC); (CP)
| | - Ataaillah Benhaddou
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Anne Camus
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Aitana Perea-Gomez
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Alice Jouneau
- Unité de Biologie du Développement et de la reproduction, UMR INRA-ENVA, INRA, Jouy-en-Josas, France
| | - Valérie Mezger
- Epigenetics and Cell Fate, UMR7216, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Francina Langa
- Centre d'Ingénierie Génétique Murine, Institut Pasteur, Paris, France
| | - Sascha Ott
- Warwick Systems Biology Centre, University of Warwick, Coventry, United Kingdom
| | - Délara Sabéran-Djoneidi
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
- Epigenetics and Cell Fate, UMR7216, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Jérôme Collignon
- Institut Jacques Monod, UMR 7592, CNRS, Université Paris-Diderot, Sorbonne Paris Cité, Paris, France
- * E-mail: (JC); (CP)
| |
Collapse
|
18
|
Boroviak T, Loos R, Bertone P, Smith A, Nichols J. The ability of inner-cell-mass cells to self-renew as embryonic stem cells is acquired following epiblast specification. Nat Cell Biol 2014; 16:516-28. [PMID: 24859004 PMCID: PMC4878656 DOI: 10.1038/ncb2965] [Citation(s) in RCA: 343] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 04/11/2014] [Indexed: 12/14/2022]
Abstract
The precise relationship of embryonic stem cells (ESCs) to cells in the mouse embryo remains controversial. We present transcriptional and functional data to identify the embryonic counterpart of ESCs. Marker profiling shows that ESCs are distinct from early inner cell mass (ICM) and closely resemble pre-implantation epiblast. A characteristic feature of mouse ESCs is propagation without ERK signalling. Single-cell culture reveals that cell-autonomous capacity to thrive when the ERK pathway is inhibited arises late during blastocyst development and is lost after implantation. The frequency of deriving clonal ESC lines suggests that all E4.5 epiblast cells can become ESCs. We further show that ICM cells from early blastocysts can progress to ERK independence if provided with a specific laminin substrate. These findings suggest that formation of the epiblast coincides with competence for ERK-independent self-renewal in vitro and consequent propagation as ESC lines.
Collapse
Affiliation(s)
- Thorsten Boroviak
- Wellcome Trust – Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK,
| | - Remco Loos
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, UK,
| | - Paul Bertone
- Wellcome Trust – Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK,
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, UK,
- Genome Biology and Developmental Biology Units, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany,
| | - Austin Smith
- Wellcome Trust – Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK,
- Department of Biochemistry, University of Cambridge, UK
| | - Jennifer Nichols
- Wellcome Trust – Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK,
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK,
| |
Collapse
|
19
|
Quantitative transcriptomics using designed primer-based amplification. Sci Rep 2014; 3:1740. [PMID: 23624976 PMCID: PMC3638165 DOI: 10.1038/srep01740] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 04/15/2013] [Indexed: 02/01/2023] Open
Abstract
We developed a novel Designed Primer-based RNA-sequencing strategy (DP-seq) that uses a defined set of heptamer primers to amplify the majority of expressed transcripts from limiting amounts of mRNA, while preserving their relative abundance. Our strategy reproducibly yielded high levels of amplification from as low as 50 picograms of mRNA while offering a dynamic range of over five orders of magnitude in RNA concentrations. We also demonstrated the potential of DP-seq to selectively suppress the amplification of the highly expressing ribosomal transcripts by more than 70% in our sequencing library. Using lineage segregation in embryonic stem cell cultures as a model of early mammalian embryogenesis, DP-seq revealed novel sets of low abundant transcripts, some corresponding to the identity of cellular progeny before they arise, reflecting the specification of cell fate prior to actual germ layer segregation.
Collapse
|
20
|
Gaunt SJ, George M, Paul YL. Direct activation of a mouse Hoxd11 axial expression enhancer by Gdf11/Smad signalling. Dev Biol 2013; 383:52-60. [PMID: 24016758 DOI: 10.1016/j.ydbio.2013.08.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 08/28/2013] [Accepted: 08/29/2013] [Indexed: 12/19/2022]
Abstract
A Hoxd11/lacZ reporter, expressed with a Hoxd11-like axial expression pattern in transgenic mouse embryos, is stimulated in tailbud fragments when cultured in presence of Gdf11, a TGF-β growth/differentiation factor. The same construct is also stimulated by Gdf11 when transiently transfected into cultures of HepG2 cells. Stimulation of the reporter in HepG2 cells is enhanced where it contains only the 332 bp Hoxd11 enhancer region VIII upstream or downstream of a luciferase or lacZ reporter. This enhancer contains three elements conserved from fish to mice, one of which has the sequence of a Smad3/4 binding element. Mutation of this motif inhibits the ability of Gdf11 to enhance reporter activity in the HepG2 cell assay. Chromatin immunoprecipitation experiments show direct evidence of Smad2/3 protein binding to the Hoxd11 region VIII enhancer. The action of Gdf11 upon Hoxd11 in HepG2 cells is inhibited, at least in part, by SIS3, a specific inhibitor of Smad3. SIS3 also produces partial inhibition of Hoxd11/lacZ expression in cultured transgenic tailbuds, indicating that Smad3 may play a similar role in the embryonic expression of Hoxd11. Transgenic mouse experiments show that the Smad binding motif is essential for the axial expression of Hoxd11/lacZ reporter in the embryo tailbud, posterior mesoderm and neurectoderm.
Collapse
Affiliation(s)
- Stephen J Gaunt
- Laboratory for Development and Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK; The Babraham Institute, Babraham, Cambridge CB22 3AT, UK.
| | | | | |
Collapse
|
21
|
TBX3 Directs Cell-Fate Decision toward Mesendoderm. Stem Cell Reports 2013; 1:248-65. [PMID: 24319661 PMCID: PMC3849240 DOI: 10.1016/j.stemcr.2013.08.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/06/2013] [Accepted: 08/07/2013] [Indexed: 12/19/2022] Open
Abstract
Cell-fate decisions and pluripotency are dependent on networks of key transcriptional regulators. Recent reports demonstrated additional functions of pluripotency-associated factors during early lineage commitment. The T-box transcription factor TBX3 has been implicated in regulating embryonic stem cell self-renewal and cardiogenesis. Here, we show that TBX3 is dynamically expressed during specification of the mesendoderm lineages in differentiating embryonic stem cells (ESCs) in vitro and in developing mouse and Xenopus embryos in vivo. Forced TBX3 expression in ESCs promotes mesendoderm specification by directly activating key lineage specification factors and indirectly by enhancing paracrine Nodal/Smad2 signaling. TBX3 loss-of-function analyses in the Xenopus underline its requirement for mesendoderm lineage commitment. Moreover, we uncovered a functional redundancy between TBX3 and Tbx2 during Xenopus gastrulation. Taken together, we define further facets of TBX3 actions and map TBX3 as an upstream regulator of the mesendoderm transcriptional program during gastrulation. T-box transcription factor TBX3 is involved in early embryonic events Key transcription factor promoters of mesendoderm formation are occupied by TBX3 TBX3 promotes mesendodermal fate of mESCs
Collapse
|
22
|
Tykwinska K, Lauster R, Knaus P, Rosowski M. Growth and differentiation factor 3 induces expression of genes related to differentiation in a model of cancer stem cells and protects them from retinoic acid-induced apoptosis. PLoS One 2013; 8:e70612. [PMID: 23950971 PMCID: PMC3741270 DOI: 10.1371/journal.pone.0070612] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 06/20/2013] [Indexed: 01/01/2023] Open
Abstract
Misexpression of growth factors, particularly those related to stem cell-like phenotype, is often observed in several cancer types. It has been found to influence parameters of disease progression like cell proliferation, differentiation, maintenance of undifferentiated phenotype and modulation of the immune system. GDF3 is a TGFB family member associated with pluripotency and differentiation during embryonic development that has been previously reported to be re-expressed in a number of cancer types. However, its role in tumor development and progression has not been clarified yet. In this study we decipher the role of GDF3 in an in vitro model of cancer stem cells, NCCIT cells. By classical approach to study protein function combined with high-throughput technique for transcriptome analysis and differentiation assays we evaluated GDF3 as a potential therapeutic target. We observed that GDF3 robustly induces a panel of genes related to differentiation, including several potent tumor suppressors, without impacting the proliferative capacity. Moreover, we report for the first time the protective effect of GDF3 against retinoic acid-induced apoptosis in cells with stem cell-like properties. Our study implies that blocking of GDF3 combined with retinoic acid-treatment of solid cancers is a compelling direction for further investigations, which can lead to re-design of cancer differentiation therapies.
Collapse
Affiliation(s)
- Karolina Tykwinska
- Institute of Medical Biotechnology, Department of Biotechnology, Technische Universität Berlin, Berlin, Germany.
| | | | | | | |
Collapse
|
23
|
Main H, Radenkovic J, Jin SB, Lendahl U, Andersson ER. Notch signaling maintains neural rosette polarity. PLoS One 2013; 8:e62959. [PMID: 23675446 PMCID: PMC3651093 DOI: 10.1371/journal.pone.0062959] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 03/26/2013] [Indexed: 12/22/2022] Open
Abstract
Formation of the metazoan body plan requires a complex interplay of morphological changes and patterning, and central to these processes is the establishment of apical/basal cell polarity. In the developing nervous system, apical/basal cell polarity is essential for neural tube closure and maintenance of the neural stem cell population. In this report we explore how a signaling pathway important for nervous system development, Notch signaling, impacts on apical/basal cell polarity in neural differentiation. CSL(-/-) mouse embryos, which are devoid of canonical Notch signaling, demonstrated a neural tube phenotype consistent with cell polarity and convergent extension defects, including deficiencies in the restricted expression of apical polarity markers in the neuroepithelium. CSL(-/-) mouse embryonic stem (ES) cells, cultured at low density, behaved as wild-type in the establishment of neural progenitors and apical specification, though progression through rosette formation, an in vitro correlate of neurulation, required CSL for correct maintenance of rosette structure and regulation of neuronal differentiation. Similarly, acute pharmacological inhibition of Notch signaling led to the breakdown of neural rosettes and accelerated neuronal differentiation. In addition to functional Notch signaling, rosette integrity was found to require actin polymerization and Rho kinase (ROCK) activity. Disruption of rosettes through inhibition of actin polymerization or ROCK activity, however, had no effect on neuronal differentiation, indicating that rosette maintenance is not a prerequisite for normal neuronal differentiation. In conclusion, our data indicate that Notch signaling plays a role not only in differentiation, but also in organization and maintenance of polarity during development of the early nervous system.
Collapse
Affiliation(s)
- Heather Main
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jelena Radenkovic
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Shao-bo Jin
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Emma R. Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
|
25
|
Redshaw N, Camps C, Sharma V, Motallebipour M, Guzman-Ayala M, Oikonomopoulos S, Thymiakou E, Ragoussis J, Episkopou V. TGF-β/Smad2/3 signaling directly regulates several miRNAs in mouse ES cells and early embryos. PLoS One 2013; 8:e55186. [PMID: 23390484 PMCID: PMC3559380 DOI: 10.1371/journal.pone.0055186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/19/2012] [Indexed: 11/30/2022] Open
Abstract
The Transforming Growth Factor-β (TGF-β) signaling pathway is one of the major pathways essential for normal embryonic development and tissue homeostasis, with anti-tumor but also pro-metastatic properties in cancer. This pathway directly regulates several target genes that mediate its downstream functions, however very few microRNAs (miRNAs) have been identified as targets. miRNAs are modulators of gene expression with essential roles in development and a clear association with diseases including cancer. Little is known about the transcriptional regulation of the primary transcripts (pri-miRNA, pri-miR) from which several mature miRNAs are often derived. Here we present the identification of miRNAs regulated by TGF-β signaling in mouse embryonic stem (ES) cells and early embryos. We used an inducible ES cell system to maintain high levels of the TGF-β activated/phosphorylated Smad2/3 effectors, which are the transcription factors of the pathway, and a specific inhibitor that blocks their activation. By performing short RNA deep-sequencing after 12 hours Smad2/3 activation and after 16 hours inhibition, we generated a database of responsive miRNAs. Promoter/enhancer analysis of a subset of these miRNAs revealed that the transcription of pri-miR-181c/d and the pri-miR-341∼3072 cluster were found to depend on activated Smad2/3. Several of these miRNAs are expressed in early mouse embryos, when the pathway is known to play an essential role. Treatment of embryos with TGF-β inhibitor caused a reduction of their levels confirming that they are targets of this pathway in vivo. Furthermore, we showed that pri-miR-341∼3072 transcription also depends on FoxH1, a known Smad2/3 transcription partner during early development. Together, our data show that miRNAs are regulated directly by the TGF-β/Smad2/3 pathway in ES cells and early embryos. As somatic abnormalities in functions known to be regulated by the TGF-β/Smad2/3 pathway underlie tumor suppression and metastasis, this research also provides a resource for miRNAs involved in cancer.
Collapse
Affiliation(s)
- Nicholas Redshaw
- Department of Medicine, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Carme Camps
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Vikas Sharma
- Department of Medicine, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Mehdi Motallebipour
- Department of Medicine, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Marcela Guzman-Ayala
- Department of Medicine, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Spyros Oikonomopoulos
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Efstathia Thymiakou
- Department of Medicine, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Jiannis Ragoussis
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Vasso Episkopou
- Department of Medicine, Division of Brain Sciences, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
26
|
Gray PC, Vale W. Cripto/GRP78 modulation of the TGF-β pathway in development and oncogenesis. FEBS Lett 2012; 586:1836-45. [PMID: 22306319 PMCID: PMC3723343 DOI: 10.1016/j.febslet.2012.01.051] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 01/24/2012] [Accepted: 01/25/2012] [Indexed: 10/14/2022]
Abstract
Cripto is a small, GPI-anchored signaling protein that regulates cellular survival, proliferation, differentiation and migration during normal developmental processes and tumorigenesis. Cripto functions as an obligatory co-receptor for the TGF-β ligands Nodal, GDF1 and GDF3 but attenuates signaling of others such as activin-A, activin-B and TGF-β1. Soluble, secreted forms of Cripto also activate Src, ras/raf/MAPK and PI3K/Akt pathways via a mechanism that remains largely obscure. This review describes the biological roles and signaling mechanisms of Cripto, highlighting our identification of the 78 kDa glucose regulated protein (GRP78) as a cell surface receptor/co-factor required for Cripto signaling via both TGF-β and Src/MAPK/PI3K pathways. We discuss emerging evidence indicating that Cripto/GRP78 signaling regulates normal somatic stem cells and their tumorigenic counterparts.
Collapse
Affiliation(s)
- Peter C Gray
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, United States.
| | | |
Collapse
|
27
|
Activin induces cortical interneuron identity and differentiation in embryonic stem cell-derived telencephalic neural precursors. Nat Commun 2012; 3:841. [PMID: 22588303 DOI: 10.1038/ncomms1817] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 03/29/2012] [Indexed: 12/15/2022] Open
Abstract
Understanding the mechanisms underlying neural progenitor differentiation and neuronal fate specification is critical for the use of embryonic stem cells (ESCs) for regenerative medicine. Cortical interneurons are of particular interest for cell transplantation; however, only a limited subset of these neurons can be generated from ESCs. Here we uncover a pivotal role for Activin in regulating the differentiation and identity of telencephalic neural precursors derived from mouse and human ESCs. We show that Activin directly inhibits the mitogenic sonic hedgehog pathway in a Gli3-dependent manner while enhancing retinoic acid signalling, the pro-neurogenic pathway. In addition, we demonstrate that Activin provides telencephalic neural precursors with positional cues that specifically promote the acquisition of a calretinin interneuron fate by controlling the expression of genes that regulate cortical interneuron identity. This work demonstrates a novel means for regulating neuronal differentiation and specification of subtype identity.
Collapse
|
28
|
Souquet B, Tourpin S, Messiaen S, Moison D, Habert R, Livera G. Nodal signaling regulates the entry into meiosis in fetal germ cells. Endocrinology 2012; 153:2466-73. [PMID: 22396454 DOI: 10.1210/en.2011-2056] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The mechanisms regulating the entry into meiosis in mammalian germ cells remain incompletely understood. We investigated the involvement of the TGF-β family members in fetal germ cell meiosis initiation. Nodal, a member of the TGF-β family, and its target genes are precociously expressed in embryonic gonads and show sexual dimorphism in favor of the developing testis. Nodal receptor genes, Acvr2a and Acvr2b, Alk4, and Tdgf1/Cripto, were identified in male germ cells. Nodal itself, Tdgf1, and Lefty1 and Lefty2 are targets of Nodal signaling and were all found specifically expressed in male germ cells. To elucidate the role of this signaling pathway, activin-like kinases that mediate TGF-β/Nodal/activin signaling were inhibited in 11.5 d postconception testis in organotypic culture. Activin-like kinases inhibition disrupted normal male germ cell development and induced germ cell entry into meiosis such as that observed in female germ cells at the equivalent stage. Interestingly Stra8, the gatekeeper of the mitotic/meiotic switch, was induced independently of any change of either Cyp26b1 or Fgf9 expression, the two genes currently identified as testicular meiotic inhibitors. On the other hand, recombinant Nodal significantly dampened Stra8 expression and germ cell meiosis in cultured 11.5 d postconception ovaries. Our results allowed us to propose for the first time an autocrine role of Nodal during the development of germ cells and indicate that members of the TGB-β family may reinforce the male fate and prevent meiosis in embryonic germ cells.
Collapse
Affiliation(s)
- Benoit Souquet
- Commissariat à l'Energie Atomique (CEA)/Direction des Science du Vivant (DSV), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Service Cellules Souches et Radiation (SCSR), Laboratoire de Développement des Gonades (LDG), Université Paris Diderot, Sorbonne Paris Cité, France
| | | | | | | | | | | |
Collapse
|
29
|
Scheubert L, Schmidt R, Repsilber D, Lustrek M, Fuellen G. Learning biomarkers of pluripotent stem cells in mouse. DNA Res 2011; 18:233-51. [PMID: 21791477 PMCID: PMC3158465 DOI: 10.1093/dnares/dsr016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2011] [Accepted: 05/10/2011] [Indexed: 01/04/2023] Open
Abstract
Pluripotent stem cells are able to self-renew, and to differentiate into all adult cell types. Many studies report data describing these cells, and characterize them in molecular terms. Machine learning yields classifiers that can accurately identify pluripotent stem cells, but there is a lack of studies yielding minimal sets of best biomarkers (genes/features). We assembled gene expression data of pluripotent stem cells and non-pluripotent cells from the mouse. After normalization and filtering, we applied machine learning, classifying samples into pluripotent and non-pluripotent with high cross-validated accuracy. Furthermore, to identify minimal sets of best biomarkers, we used three methods: information gain, random forests and a wrapper of genetic algorithm and support vector machine (GA/SVM). We demonstrate that the GA/SVM biomarkers work best in combination with each other; pathway and enrichment analyses show that they cover the widest variety of processes implicated in pluripotency. The GA/SVM wrapper yields best biomarkers, no matter which classification method is used. The consensus best biomarker based on the three methods is Tet1, implicated in pluripotency just recently. The best biomarker based on the GA/SVM wrapper approach alone is Fam134b, possibly a missing link between pluripotency and some standard surface markers of unknown function processed by the Golgi apparatus.
Collapse
Affiliation(s)
- Lena Scheubert
- Institute of Computer Science, University of Osnabrück, Germany
| | | | | | | | | |
Collapse
|
30
|
Lee KL, Lim SK, Orlov YL, Yit LY, Yang H, Ang LT, Poellinger L, Lim B. Graded Nodal/Activin signaling titrates conversion of quantitative phospho-Smad2 levels into qualitative embryonic stem cell fate decisions. PLoS Genet 2011; 7:e1002130. [PMID: 21731500 PMCID: PMC3121749 DOI: 10.1371/journal.pgen.1002130] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 05/01/2011] [Indexed: 12/11/2022] Open
Abstract
Nodal and Activin are morphogens of the TGFbeta superfamily of signaling molecules that direct differential cell fate decisions in a dose- and distance-dependent manner. During early embryonic development the Nodal/Activin pathway is responsible for the specification of mesoderm, endoderm, node, and mesendoderm. In contradiction to this drive towards cellular differentiation, the pathway also plays important roles in the maintenance of self-renewal and pluripotency in embryonic and epiblast stem cells. The molecular basis behind stem cell interpretation of Nodal/Activin signaling gradients and the undertaking of disparate cell fate decisions remains poorly understood. Here, we show that any perturbation of endogenous signaling levels in mouse embryonic stem cells leads to their exit from self-renewal towards divergent differentiation programs. Increasing Nodal signals above basal levels by direct stimulation with Activin promotes differentiation towards the mesendodermal lineages while repression of signaling with the specific Nodal/Activin receptor inhibitor SB431542 induces trophectodermal differentiation. To address how quantitative Nodal/Activin signals are translated qualitatively into distinct cell fates decisions, we performed chromatin immunoprecipitation of phospho-Smad2, the primary downstream transcriptional factor of the Nodal/Activin pathway, followed by massively parallel sequencing, and show that phospho-Smad2 binds to and regulates distinct subsets of target genes in a dose-dependent manner. Crucially, Nodal/Activin signaling directly controls the Oct4 master regulator of pluripotency by graded phospho-Smad2 binding in the promoter region. Hence stem cells interpret and carry out differential Nodal/Activin signaling instructions via a corresponding gradient of Smad2 phosphorylation that selectively titrates self-renewal against alternative differentiation programs by direct regulation of distinct target gene subsets and Oct4 expression.
Collapse
Affiliation(s)
- Kian Leong Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology, and Research, Singapore, Singapore
- * E-mail: (KLL); –star.edu.sg (BL)
| | - Sandy Keat Lim
- Genome Institute of Singapore, Agency for Science, Technology, and Research, Singapore, Singapore
- Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Yuriy Lvovich Orlov
- Genome Institute of Singapore, Agency for Science, Technology, and Research, Singapore, Singapore
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Le Yau Yit
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Henry Yang
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Lay Teng Ang
- Genome Institute of Singapore, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Lorenz Poellinger
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Bing Lim
- Genome Institute of Singapore, Agency for Science, Technology, and Research, Singapore, Singapore
- Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (KLL); –star.edu.sg (BL)
| |
Collapse
|
31
|
Ghosh P, Garcia-Marcos M, Farquhar MG. GIV/Girdin is a rheostat that fine-tunes growth factor signals during tumor progression. Cell Adh Migr 2011; 5:237-48. [PMID: 21546796 DOI: 10.4161/cam.5.3.15909] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
GIV/Girdin is a multidomain signaling molecule that enhances PI3K-Akt signals downstream of both G protein-coupled and growth factor receptors. We previously reported that GIV triggers cell migration via its C-terminal guanine-nucleotide exchange factor (GEF) motif that activates Gαi. Recently we discovered that GIV's C-terminus directly interacts with the epidermal growth factor receptor (EGFR), and when its GEF function is intact, a Gαi-GIV-EGFR signaling complex assembles. By coupling G proteins to growth factor receptors, GIV is uniquely poised to intercept the incoming receptor-initiated signals and modulate them via G protein intermediates. Subsequent work has revealed that expression of the highly specialized C-terminus of GIV undergoes a bipartite dysregulation during oncogenesis-full length GIV with an intact C-terminus is expressed at levels ~20-50-fold above normal in highly invasive cancer cells and metastatic tumors, but its C-terminus is truncated by alternative splicing in poorly invasive cancer cells and non-invasive tumors. The consequences of such dysregulation on graded signal transduction and cellular phenotypes in the normal epithelium and its implication during tumor progression are discussed herein. Based on the fact that GIV grades incoming signals initiated by ligand-activated receptors by linking them to cyclical activation of G proteins, we propose that GIV is a molecular rheostat for signal transduction.
Collapse
Affiliation(s)
- Pradipta Ghosh
- Department of Medicine, School of Medicine, University of California-San Diego, La Jolla, CA, USA.
| | | | | |
Collapse
|
32
|
Galvin-Burgess KE, Vivian JL. Transforming growth factor-beta superfamily in mouse embryonic stem cell self-renewal. VITAMINS AND HORMONES 2011; 87:341-65. [PMID: 22127250 DOI: 10.1016/b978-0-12-386015-6.00035-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Embryonic stem (ES) cells are pluripotent cells that maintain the capability of undifferentiated self-renewal in culture. As mouse ES cells have the capacity to give rise to all the tissues of the body, they are an excellent developmental biology model system and a model for regenerative therapies. The extracellular cues and the intracellular signaling cascades that regulate ES cell self-renewal and cell-fate choices are complex and actively studied. Many developmental signaling pathways regulate the ES cell phenotype, and their intracellular programs interact to modulate the gene networks controlling ES cell pluripotency. This review focuses on the current understanding and outstanding questions of the roles of the transforming growth factor-beta-related signaling pathways in regulating pluripotency and differentiation of mouse ES cells. The complex dichotomic roles of bone morphogenetic protein signaling in maintaining the undifferentiated state and also inducing specific cell fates will be reviewed. The emerging roles of Nodal signaling in ES cell self-renewal will also be discussed.
Collapse
Affiliation(s)
- Katherine E Galvin-Burgess
- Department of Pathology and Laboratory Medicine, Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, USA
| | | |
Collapse
|
33
|
Smad2 mediates Activin/Nodal signaling in mesendoderm differentiation of mouse embryonic stem cells. Cell Res 2010; 20:1306-18. [PMID: 21079647 DOI: 10.1038/cr.2010.158] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Although Activin/Nodal signaling regulates pluripotency of human embryonic stem (ES) cells, how this signaling acts in mouse ES cells remains largely unclear. To investigate this, we confirmed that mouse ES cells possess active Smad2-mediated Activin/Nodal signaling and found that Smad2-mediated Activin/Nodal signaling is dispensable for self-renewal maintenance but is required for proper differentiation toward the mesendoderm lineage. To gain insights into the underlying mechanisms, Smad2-associated genes were identified by genome-wide chromatin immunoprecipitation-chip analysis. The results showed that there is a transcriptional correlation between Smad2 binding and Activin/Nodal signaling modulation, and that the development-related genes were enriched among the Smad2-bound targets. We further identified Tapbp as a key player in mesendoderm differentiation of mouse ES cells acting downstream of the Activin/Nodal-Smad2 pathway. Taken together, our findings suggest that Smad2-mediated Activin/Nodal signaling orchestrates mesendoderm lineage commitment of mouse ES cells through direct modulation of corresponding developmental regulator expression.
Collapse
|
34
|
Abstract
Nodal signals belong to the TGF-beta superfamily and are essential for the induction of mesoderm and endoderm and the determination of the left-right axis. Nodal signals can act as morphogens-they have concentration-dependent effects and can act at a distance from their source of production. Nodal and its feedback inhibitor Lefty form an activator/inhibitor pair that behaves similarly to postulated reaction-diffusion models of tissue patterning. Nodal morphogen activity is also regulated by microRNAs, convertases, TGF-beta signals, coreceptors, and trafficking factors. This article describes how Nodal morphogens pattern embryonic fields and discusses how Nodal morphogen signaling is modulated.
Collapse
|
35
|
Galvin KE, Travis ED, Yee D, Magnuson T, Vivian JL. Nodal signaling regulates the bone morphogenic protein pluripotency pathway in mouse embryonic stem cells. J Biol Chem 2010; 285:19747-56. [PMID: 20427282 DOI: 10.1074/jbc.m109.077347] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Members of the transforming growth factor-beta superfamily play essential roles in both the pluripotency and differentiation of embryonic stem (ES) cells. Although bone morphogenic proteins (BMPs) maintain pluripotency of undifferentiated mouse ES cells, the role of autocrine Nodal signaling is less clear. Pharmacological, molecular, and genetic methods were used to further understand the roles and potential interactions of these pathways. Treatment of undifferentiated ES cells with SB431542, a pharmacological inhibitor of Smad2 signaling, resulted in a rapid reduction of phosphorylated Smad2 and altered the expression of several putative downstream targets. Unexpectedly, inhibition of the Nodal signaling pathway resulted in enhanced BMP signaling, as assessed by Smad1/5 phosphorylation. SB431542-treated cells also demonstrated significant induction of the Id genes, which are known direct targets of BMP signaling and important factors in ES cell pluripotency. Inhibition of BMP signaling decreased the SB431542-mediated phosphorylation of Smad1/5 and induction of Id genes, suggesting that BMP signaling is necessary for some Smad2-mediated activity. Because Smad7, a known inhibitory factor to both Nodal and BMP signaling, was down-regulated following inhibition of Nodal-Smad2 signaling, the contribution of Smad7 to the cross-talk between the transforming growth factor-beta pathways in ES cells was examined. Biochemical manipulation of Smad7 expression, through shRNA knockdown or inducible gene expression, significantly reduced the SB431542-mediated phosphorylation of Smad1/5 and induction of the Id genes. We conclude that autocrine Nodal signaling in undifferentiated mouse ES cells modulates the vital pluripotency pathway of BMP signaling.
Collapse
Affiliation(s)
- Katherine E Galvin
- Department of Pathology and Laboratory Medicine and Division of Cancer and Developmental Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | |
Collapse
|
36
|
Pan D, Zhu Q, Luo K. SnoN functions as a tumour suppressor by inducing premature senescence. EMBO J 2009; 28:3500-13. [PMID: 19745809 PMCID: PMC2782089 DOI: 10.1038/emboj.2009.250] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 07/29/2009] [Indexed: 01/08/2023] Open
Abstract
SnoN represses TGF-beta signalling to promote cell proliferation and has been defined as a proto-oncogene partly due to its elevated expression in many human cancer cells. Although the anti-tumourigenic activity of SnoN has been suggested, the molecular basis for this has not been defined. We showed here that high levels of SnoN exert anti-oncogenic activity by inducing senescence. SnoN interacts with the promyelocytic leukaemia (PML) protein and is recruited to the PML nuclear bodies where it stabilizes p53, leading to premature senescence. Furthermore, overexpression of SnoN inhibits oncogenic transformation induced by Ras and Myc in vitro and significantly blocks papilloma development in vivo in a carcinogen-induced skin tumourigenesis model. The few papillomas that were developed displayed high levels of senescence and spontaneously regressed. Our study has revealed a novel Smad-independent pathway of SnoN function that mediates its anti-oncogenic activity.
Collapse
Affiliation(s)
- Deng Pan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
37
|
Vasilaki E, Papadimitriou E, Tajadura V, Ridley AJ, Stournaras C, Kardassis D. Transcriptional regulation of the small GTPase RhoB gene by TGFß‐induced signaling pathways. FASEB J 2009; 24:891-905. [DOI: 10.1096/fj.09-134742] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Eleftheria Vasilaki
- Department of BiochemistrySchool of MedicineUniversity of CreteHeraklionGreece
| | - Elsa Papadimitriou
- Department of BiochemistrySchool of MedicineUniversity of CreteHeraklionGreece
| | - Virginia Tajadura
- Randall Division of Cell and Molecular BiophysicsGuy's CampusKing's College LondonLondonUK
| | - Anne J. Ridley
- Randall Division of Cell and Molecular BiophysicsGuy's CampusKing's College LondonLondonUK
| | - Christos Stournaras
- Department of BiochemistrySchool of MedicineUniversity of CreteHeraklionGreece
| | - Dimitris Kardassis
- Department of BiochemistrySchool of MedicineUniversity of CreteHeraklionGreece
| |
Collapse
|
38
|
Abstract
Bone healing is a predictable process that has a high rate of success. For some patients, and in certain clinical settings, this process can be delayed or completely inhibited. This leads to significant morbidity and may also result in time lost from work, costs related to prolonged medical treatment, and continued pain at the site of nonunion or failed spinal fusion. Several growth factors, specifically BMP-2 and BMP-7, have been approved in several countries for specific indications. The use of these products and potential complications of their use are reviewed.
Collapse
Affiliation(s)
- T William Axelrad
- Department of Orthopaedic Surgery, Boston University Medical Center, Boston, MA, United States
| | | |
Collapse
|
39
|
Seuntjens E, Umans L, Zwijsen A, Sampaolesi M, Verfaillie CM, Huylebroeck D. Transforming Growth Factor type beta and Smad family signaling in stem cell function. Cytokine Growth Factor Rev 2009; 20:449-58. [PMID: 19892581 DOI: 10.1016/j.cytogfr.2009.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ligands of the Transforming Growth Factor type beta (TGFbeta) family exert multiple and sometimes opposite effects on most cell types in vivo depending on cellular context, which mainly includes the stage of the target cell, the local environment of this cell or niche, and the identity and the dosage of the ligand. Significant progress has been made in the molecular dissection of the regulation of the activity of the ligands and their intracellular signal transduction pathways, including via the canonical Smad pathway where Smads interact with many transcription factors. This knowledge together with results from functional studies within the embryology and stem cell research fields is giving us insight in the role of individual ligands and other components of this signaling system and where and how it regulates many properties of embryonic and adult stem/progenitor cells, which is anticipated to contribute to successful cell-based therapy in the future. We review and discuss recent progress on the effects of Nodal/Activin and Bone Morphogenetic Proteins (BMPs) and their canonical signaling in cells with stem cell properties. We focus on embryonic stem cells and their maintenance and pluripotency, and conversion into selected cell types of neuroectoderm, mesoderm and endoderm, on induced pluripotent cells and on neurogenic cells in the adult brain.
Collapse
Affiliation(s)
- Eve Seuntjens
- Laboratory of Molecular Biology (Celgen) of the Center for Human Genetics, University of Leuven, Flanders Institute of Biotechnology (VIB), Campus Gasthuisberg, B-3000 Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|