551
|
Abstract
Formation of a eutherian mammal requires concurrent establishment of embryonic and extraembryonic lineages. The functions of the trophectoderm and primitive endoderm are to enable implantation in the maternal uterus, axis specification and delivery of nutrients. The pluripotent epiblast represents the founding cell population of the embryo proper, which is protected from ectopic and premature differentiation until it is required to respond to inductive cues to form the fetus. While positional information plays a major role in specifying the trophoblast lineage, segregation of primitive endoderm from epiblast depends upon gradual acquisition of transcriptional identity, directed but not initiated by fibroblast growth factor (FGF) signalling. Following early cleavage divisions and formation of the blastocyst, cells of the inner cell mass lose totipotency. Developing epiblast cells transiently attain the state of naive pluripotency and competence to self-renew in vitro as embryonic stem cells and in vivo by means of diapause. This property is lost after implantation as the epiblast epithelializes and becomes primed in preparation for gastrulation and subsequent organogenesis.
Collapse
Affiliation(s)
- Thorsten Boroviak
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Jennifer Nichols
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
552
|
Sheng X, Li WB, Wang DL, Chen KH, Cao JJ, Luo Z, He J, Li MC, Liu WJ, Yu C. YAP is closely correlated with castration-resistant prostate cancer, and downregulation of YAP reduces proliferation and induces apoptosis of PC-3 cells. Mol Med Rep 2015; 12:4867-76. [PMID: 26126522 PMCID: PMC4581809 DOI: 10.3892/mmr.2015.4005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 05/19/2015] [Indexed: 11/06/2022] Open
Abstract
Yes-associated protein 65 (YAP65) has been implicated as an oncogene, and its expression is increased in human cancer. Previous studies have demonstrated that alterations in YAP activity may result in tumourigenesis of the prostate. With androgen deprivation therapies becoming progressively ineffective, often leading to life‑threatening androgen‑resistant prostate cancer (CRPC). The present study aimed to analyse the role of YAP in prostate cancer (PCa), particularly in CRPC. YAP protein was detected using immunohistochemistry and western blot analysis in different prostatic tissues. In addition, three specific RNA interference vectors targeting the human YAP gene were synthesised, and PC‑3 cells with a stable inhibition of YAP were obtained by transfection. MTT, flow cytometry, reverse transcription‑quantitative polymerase chain reaction and western blot assays were used to analyse the effects of YAP inhibition on the proliferation and apoptosis of PC‑3 cells. The frequency of cells that were positive for YAP protein in PCa (78.13%) was significantly higher, compared with para‑PCa (26.67%; P=0.007) and benign prostatic hyperplasia (0%; P=0.002). The frequency of cells, which were positive for the expression of YAP exhibited a positive correlation (P=0.008) with the Gleason score, the tumour‑node‑metastasis staging (P=0.033) and the level of prostate specific antigens (P=0.0032) in PCa. The proliferative capacity of the transfected group was significantly lower, compared with the negative control group (P=0.022). The cell‑cycle of the transfected group was arrested in the G1 stage, which was detected using flow cytometry, and there was a significant increase in the apoptosis of cells in the transfected group (P=0.002). The mRNA and protein levels of TEA domain family member 1 were inhibited in the transfected group (P=0.001 and P=0.00, respectively). Therefore, it was concluded that gene transcription and protein expression of YAP may be involved in the development of PCa, particularly CRPC, and may be a novel biomarker for investigation of the occurrence and progression of CRPC. However, the mechanism underlying the modulation of YAP in CRPC remains to be fully elucidated.
Collapse
Affiliation(s)
- Xia Sheng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wen-Bin Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - De-Lin Wang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ke-Hong Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jian-Jia Cao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhao Luo
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jiang He
- Department of Urology, University‑Town Hospital of Chongqing Medical University, Chongqing 401331, P.R. China
| | - Mei-Cai Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wu-Jiang Liu
- Department of Urology, Institute of Urology, First Hospital of Peking University, Beijing 100034, P.R. China
| | - Chao Yu
- Department of Surgery, Life Science Research Institute of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
553
|
Bedzhov I, Graham SJL, Leung CY, Zernicka-Goetz M. Developmental plasticity, cell fate specification and morphogenesis in the early mouse embryo. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0538. [PMID: 25349447 PMCID: PMC4216461 DOI: 10.1098/rstb.2013.0538] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A critical point in mammalian development is when the early embryo implants into its mother's uterus. This event has historically been difficult to study due to the fact that it occurs within the maternal tissue and therefore is hidden from view. In this review, we discuss how the mouse embryo is prepared for implantation and the molecular mechanisms involved in directing and coordinating this crucial event. Prior to implantation, the cells of the embryo are specified as precursors of future embryonic and extra-embryonic lineages. These preimplantation cell fate decisions rely on a combination of factors including cell polarity, position and cell–cell signalling and are influenced by the heterogeneity between early embryo cells. At the point of implantation, signalling events between the embryo and mother, and between the embryonic and extraembryonic compartments of the embryo itself, orchestrate a total reorganization of the embryo, coupled with a burst of cell proliferation. New developments in embryo culture and imaging techniques have recently revealed the growth and morphogenesis of the embryo at the time of implantation, leading to a new model for the blastocyst to egg cylinder transition. In this model, pluripotent cells that will give rise to the fetus self-organize into a polarized three-dimensional rosette-like structure that initiates egg cylinder formation.
Collapse
Affiliation(s)
- Ivan Bedzhov
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Sarah J L Graham
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Chuen Yan Leung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Magdalena Zernicka-Goetz
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
554
|
Hansen CG, Moroishi T, Guan KL. YAP and TAZ: a nexus for Hippo signaling and beyond. Trends Cell Biol 2015; 25:499-513. [PMID: 26045258 DOI: 10.1016/j.tcb.2015.05.002] [Citation(s) in RCA: 443] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 05/02/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
Abstract
The Hippo pathway is a potent regulator of cellular proliferation, differentiation, and tissue homeostasis. Here we review the regulatory mechanisms of the Hippo pathway and discuss the function of Yes-associated protein (YAP)/transcriptional coactivator with a PDZ-binding domain (TAZ), the prime mediators of the Hippo pathway, in stem cell biology and tissue regeneration. We highlight their activities in both the nucleus and the cytoplasm and discuss their role as a signaling nexus and integrator of several other prominent signaling pathways such as the Wnt, G protein-coupled receptor (GPCR), epidermal growth factor (EGF), bone morphogenetic protein (BMP)/transforming growth factor beta (TGFβ), and Notch pathways.
Collapse
Affiliation(s)
- Carsten Gram Hansen
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.,Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Toshiro Moroishi
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.,Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kun-Liang Guan
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.,Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
555
|
Togashi K, Kumagai J, Sato E, Shirasawa H, Shimoda Y, Makino K, Sato W, Kumazawa Y, Omori Y, Terada Y. Dysfunction in gap junction intercellular communication induces aberrant behavior of the inner cell mass and frequent collapses of expanded blastocysts in mouse embryos. J Assist Reprod Genet 2015; 32:969-76. [PMID: 25917498 PMCID: PMC4491087 DOI: 10.1007/s10815-015-0479-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 04/08/2015] [Indexed: 10/23/2022] Open
Abstract
PURPOSE We investigated the role of gap junctions (GJs) in embryological differentiation, and observed the morphological behavior of the inner cell mass (ICM) by time-lapse movie observation (TLM) with gap junction inhibitors (GJis). METHODS ICR mouse embryos were exposed to two types of GJis in CZB medium: oleamide (0 to 50 μM) and 1-heptanol (0 to 10 mM). We compared the rate of blastocyst formation at embryonic day 4.5 (E4.5) with E5.5. We also observed and evaluated the times from the second cleavage to each embryonic developing stage by TLM. We investigated embryonic distribution of DNA, Nanog protein, and Connexin 43 protein with immunofluorescent staining. RESULTS In the comparison of E4.5 with E5.5, inhibition of gap junction intercellular communication (GJIC) delayed embryonic blastocyst formation. The times from the second cleavage to blastocyst formation were significantly extended in the GJi-treated embryos (control vs with oleamide, 2224 ± 179 min vs 2354 ± 278 min, p = 0.013). Morphological differences were traced in control versus GJi-treated embryos until the hatching stage. Oleamide induced frequent severe collapses of expanded blastocysts (77.4 % versus 26.3 %, p = 0.0001) and aberrant ICM divisions connected to sticky strands (74.3 % versus 5.3 %, p = 0.0001). Immunofluorescent staining indicated Nanog-positive cells were distributed in each divided ICM. CONCLUSIONS GJIC plays an important role in blastocyst formation, collapses of expanded blastocysts, and the ICM construction in mouse embryos.
Collapse
Affiliation(s)
- Kazue Togashi
- Department of Obstetrics and Genecology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
556
|
Frum T, Ralston A. Cell signaling and transcription factors regulating cell fate during formation of the mouse blastocyst. Trends Genet 2015; 31:402-10. [PMID: 25999217 DOI: 10.1016/j.tig.2015.04.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/05/2015] [Accepted: 04/07/2015] [Indexed: 11/17/2022]
Abstract
The first cell fate decisions during mammalian development establish tissues essential for healthy pregnancy. The mouse has served as a valuable model for discovering pathways regulating the first cell fate decisions because of the ease with which early embryos can be recovered and the availability of an arsenal of classical and emerging methods for manipulating gene expression. We summarize the major pathways that govern the first cell fate decisions in mouse development. This knowledge serves as a paradigm for exploring how emergent properties of a self-organizing system can dynamically regulate gene expression and cell fate plasticity. Moreover, it brings to light the processes that establish healthy pregnancy and ES cells. We also describe unsolved mysteries and new technologies that could help to overcome experimental challenges in the field.
Collapse
Affiliation(s)
- Tristan Frum
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Amy Ralston
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
557
|
Choi I, Dasari A, Kim NH, Campbell KHS. Effects of prolonged exposure of mouse embryos to elevated temperatures on embryonic developmental competence. Reprod Biomed Online 2015; 31:171-9. [PMID: 26093856 DOI: 10.1016/j.rbmo.2015.04.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 04/30/2015] [Accepted: 04/30/2015] [Indexed: 01/15/2023]
Abstract
To investigate effects of heat stress on developmental competence, in-vitro fertilized zygotes were incubated at different temperatures until 96 h post human chorionic gonadotrophin (HCG). Under severe and moderate conditions (41°C and 40°C), most embryos did not overcome the 2-cell block. In long-term mild heat stress (39°C until 96 h post HCG), cleavage and blastocyst formation were comparable to non-heat-stress control, but the number of live pups per transferred embryo and mean litter size were significantly affected (P < 0.05) in the mild-heat-stress group (19.4%, and 5.1 ± 0.4, respectively), compared with control (41.7% and 8.3 ± 0.3, respectively). To elucidate the different competence, gene expression was examined and the numbers of inner cell mass (ICM) and trophectoderm (TE) cells were counted. Aberrant expression of genes for embryonic viability and trophoblast differentiation in the mild-heat-stressed blastocysts was found. Moreover, the expanded blastocysts in the heat-stressed group and the control had a ICM:TE ratio of 1:2.47 and 1:2.96 with average total cell numbers of 59.21 ± 2.38 and 72.79 ± 2.40, respectively (P < 0.05), indicating lower cell numbers in TE. These findings underscore that prevention of heat stress in early embryos is important for maintaining embryo viability embryos during pregnancy.
Collapse
Affiliation(s)
- Inchul Choi
- Animal Development and Biotechnology Group, School of Biosciences, The University of Nottingham, Sutton Bonington Campus, Loughborough, UK; Department of Animal Biosystem Sciences, College of Agriculture and Life Sciences, Chungnam National University 305-764, Republic of Korea.
| | - Amarnath Dasari
- Animal Development and Biotechnology Group, School of Biosciences, The University of Nottingham, Sutton Bonington Campus, Loughborough, UK; Taconic Farms Inc., Five University Place Rensselaer, NY 12144-3439, USA
| | - Nam-Hyung Kim
- Department of Animal Sciences, Chungbuk National University, Gaesin-dong, Cheongju, Chungbuk. 361-763, Republic of Korea
| | - Keith H S Campbell
- Animal Development and Biotechnology Group, School of Biosciences, The University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| |
Collapse
|
558
|
Sasaki H. Position- and polarity-dependent Hippo signaling regulates cell fates in preimplantation mouse embryos. Semin Cell Dev Biol 2015; 47-48:80-7. [PMID: 25986053 DOI: 10.1016/j.semcdb.2015.05.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 05/08/2015] [Accepted: 05/08/2015] [Indexed: 11/25/2022]
Abstract
During the preimplantation stage, mouse embryos establish two cell lineages by the time of early blastocyst formation: the trophectoderm (TE) and the inner cell mass (ICM). Historical models have proposed that the establishment of these two lineages depends on the cell position within the embryo (e.g., the positional model) or cell polarization along the apicobasal axis (e.g., the polarity model). Recent findings have revealed that the Hippo signaling pathway plays a central role in the cell fate-specification process: active and inactive Hippo signaling in the inner and outer cells promote ICM and TE fates, respectively. Intercellular adhesion activates, while apicobasal polarization suppresses Hippo signaling, and a combination of these processes determines the spatially regulated activation of the Hippo pathway in 32-cell-stage embryos. Therefore, there is experimental evidence in favor of both positional and polarity models. At the molecular level, phosphorylation of the Hippo-pathway component angiomotin at adherens junctions (AJs) in the inner (apolar) cells activates the Lats protein kinase and triggers Hippo signaling. In the outer cells, however, cell polarization sequesters Amot from basolateral AJs and suppresses activation of the Hippo pathway. Other mechanisms, including asymmetric cell division and Notch signaling, also play important roles in the regulation of embryonic development. In this review, I discuss how these mechanisms cooperate with the Hippo signaling pathway during cell fate-specification processes.
Collapse
Affiliation(s)
- Hiroshi Sasaki
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
559
|
Wittkorn E, Sarkar A, Garcia K, Kango-Singh M, Singh A. The Hippo pathway effector Yki downregulates Wg signaling to promote retinal differentiation in the Drosophila eye. Development 2015; 142:2002-13. [PMID: 25977365 DOI: 10.1242/dev.117358] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 04/16/2015] [Indexed: 01/22/2023]
Abstract
The evolutionarily conserved Hippo signaling pathway is known to regulate cell proliferation and maintain tissue homeostasis during development. We found that activation of Yorkie (Yki), the effector of the Hippo signaling pathway, causes separable effects on growth and differentiation of the Drosophila eye. We present evidence supporting a role for Yki in suppressing eye fate by downregulation of the core retinal determination genes. Other upstream regulators of the Hippo pathway mediate this effect of Yki on retinal differentiation. Here, we show that, in the developing eye, Yki can prevent retinal differentiation by blocking morphogenetic furrow (MF) progression and R8 specification. The inhibition of MF progression is due to ectopic induction of Wingless (Wg) signaling and Homothorax (Hth), the negative regulators of eye development. Modulating Wg signaling can modify Yki-mediated suppression of eye fate. Furthermore, ectopic Hth induction due to Yki activation in the eye is dependent on Wg. Last, using Cut (Ct), a marker for the antennal fate, we show that suppression of eye fate by hyperactivation of yki does not change the cell fate (from eye to antenna-specific fate). In summary, we provide the genetic mechanism by which yki plays a role in cell fate specification and differentiation - a novel aspect of Yki function that is emerging from multiple model organisms.
Collapse
Affiliation(s)
- Erika Wittkorn
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | - Ankita Sarkar
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | - Kristine Garcia
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | - Madhuri Kango-Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA Premedical Program, University of Dayton, Dayton, OH 45469, USA Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH 45469, USA
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA Premedical Program, University of Dayton, Dayton, OH 45469, USA Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH 45469, USA
| |
Collapse
|
560
|
Lorthongpanich C, Issaragrisil S. Emerging Role of the Hippo Signaling Pathway in Position Sensing and Lineage Specification in Mammalian Preimplantation Embryos. Biol Reprod 2015; 92:143. [PMID: 25947059 DOI: 10.1095/biolreprod.114.127803] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/29/2015] [Indexed: 12/29/2022] Open
Abstract
In preimplantation mouse embryos, the first lineage differentiation takes place in the 8- to 16-cell-stage embryo and results in formation of the trophectoderm (TE) and inner cell mass (ICM), which will give rise to the trophoblast of the placenta and the embryo proper, respectively. Although, it is widely accepted that positioning of a cell within the embryo influences lineage differentiation, the mechanism underlying differential lineage differentiation and how it involves cell position are largely unknown. Interestingly, novel cues from the Hippo pathway have been recently demonstrated in the preimplantation mouse embryo. Unlike the mechanisms reported from epithelium-cultured cells, the Hippo pathway was found to be responsible for translating positional information to lineage specification through a position-sensing mechanism. Disruption of Hippo pathway-component genes in early embryos results in failure of lineage specification and failure of postimplantation development. In this review, we discuss the unique role of the Hippo signaling pathway in early embryo development and its role in lineage specification. Understanding the activity and regulation of the Hippo pathway may offer new insights into other areas of developmental biology that evolve from understanding of this cell-fate specification in the early embryonic cell.
Collapse
Affiliation(s)
- Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
561
|
Cao Z, Carey TS, Ganguly A, Wilson CA, Paul S, Knott JG. Transcription factor AP-2γ induces early Cdx2 expression and represses HIPPO signaling to specify the trophectoderm lineage. Development 2015; 142:1606-15. [PMID: 25858457 DOI: 10.1242/dev.120238] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/06/2015] [Indexed: 01/31/2023]
Abstract
Cell fate decisions are fundamental to the development of multicellular organisms. In mammals the first cell fate decision involves segregation of the pluripotent inner cell mass and the trophectoderm, a process regulated by cell polarity proteins, HIPPO signaling and lineage-specific transcription factors such as CDX2. However, the regulatory mechanisms that operate upstream to specify the trophectoderm lineage have not been established. Here we report that transcription factor AP-2γ (TFAP2C) functions as a novel upstream regulator of Cdx2 expression and position-dependent HIPPO signaling in mice. Loss- and gain-of-function studies and promoter analysis revealed that TFAP2C binding to an intronic enhancer is required for activation of Cdx2 expression during early development. During the 8-cell to morula transition TFAP2C potentiates cell polarity to suppress HIPPO signaling in the outside blastomeres. TFAP2C depletion triggered downregulation of PARD6B, loss of apical cell polarity, disorganization of F-actin, and activation of HIPPO signaling in the outside blastomeres. Rescue experiments using Pard6b mRNA restored cell polarity but only partially corrected position-dependent HIPPO signaling, suggesting that TFAP2C negatively regulates HIPPO signaling via multiple pathways. Several genes involved in regulation of the actin cytoskeleton (including Rock1, Rock2) were downregulated in TFAP2C-depleted embryos. Inhibition of ROCK1 and ROCK2 activity during the 8-cell to morula transition phenocopied TFAP2C knockdown, triggering a loss of position-dependent HIPPO signaling and decrease in Cdx2 expression. Altogether, these results demonstrate that TFAP2C facilitates trophectoderm lineage specification by functioning as a key regulator of Cdx2 transcription, cell polarity and position-dependent HIPPO signaling.
Collapse
Affiliation(s)
- Zubing Cao
- Developmental Epigenetics Laboratory, Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Timothy S Carey
- Developmental Epigenetics Laboratory, Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Avishek Ganguly
- Department of Pathology and Laboratory Medicine, Institute of Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Catherine A Wilson
- Developmental Epigenetics Laboratory, Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine, Institute of Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jason G Knott
- Developmental Epigenetics Laboratory, Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
562
|
Wang C, Chen Y, Deng H, Gao S, Li L. Rbm46 Regulates Trophectoderm Differentiation by Stabilizing Cdx2 mRNA in Early Mouse Embryos. Stem Cells Dev 2015; 24:904-15. [DOI: 10.1089/scd.2014.0323] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Chenchen Wang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University Stem Cell Research Center, China National Center for International Research, Peking University Health Science Center, Beijing, China
- SARI Center for Stem Cell and Nanomedicine, Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Shanghai, China
| | - Yuanfan Chen
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University Stem Cell Research Center, China National Center for International Research, Peking University Health Science Center, Beijing, China
- SARI Center for Stem Cell and Nanomedicine, Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Shanghai, China
| | - Hongkui Deng
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University Stem Cell Research Center, China National Center for International Research, Peking University Health Science Center, Beijing, China
| | - Shaorong Gao
- National Institute of Biological Sciences, Beijing, China
| | - Lingsong Li
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University Stem Cell Research Center, China National Center for International Research, Peking University Health Science Center, Beijing, China
- SARI Center for Stem Cell and Nanomedicine, Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
563
|
Santucci M, Vignudelli T, Ferrari S, Mor M, Scalvini L, Bolognesi ML, Uliassi E, Costi MP. The Hippo Pathway and YAP/TAZ-TEAD Protein-Protein Interaction as Targets for Regenerative Medicine and Cancer Treatment. J Med Chem 2015; 58:4857-73. [PMID: 25719868 DOI: 10.1021/jm501615v] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Hippo pathway is an important organ size control signaling network and the major regulatory mechanism of cell-contact inhibition. Yes associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are its targets and terminal effectors: inhibition of the pathway promotes YAP/TAZ translocation to the nucleus, where they interact with transcriptional enhancer associate domain (TEAD) transcription factors and coactivate the expression of target genes, promoting cell proliferation. Defects in the pathway can result in overgrowth phenotypes due to deregulation of stem-cell proliferation and apoptosis; members of the pathway are directly involved in cancer development. The pharmacological regulation of the pathway might be useful in cancer prevention, treatment, and regenerative medicine applications; currently, a few compounds can selectively modulate the pathway. In this review, we present an overview of the Hippo pathway, the sequence and structural analysis of YAP/TAZ, the known pharmacological modulators of the pathway, especially those targeting YAP/TAZ-TEAD interaction.
Collapse
Affiliation(s)
- Matteo Santucci
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| | - Tatiana Vignudelli
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| | - Stefania Ferrari
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| | - Marco Mor
- ‡Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Laura Scalvini
- ‡Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Maria Laura Bolognesi
- §Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Elisa Uliassi
- §Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Maria Paola Costi
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| |
Collapse
|
564
|
Blij S, Parenti A, Tabatabai-Yazdi N, Ralston A. Cdx2 efficiently induces trophoblast stem-like cells in naïve, but not primed, pluripotent stem cells. Stem Cells Dev 2015; 24:1352-65. [PMID: 25625326 DOI: 10.1089/scd.2014.0395] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Diverse pluripotent stem cell lines have been derived from the mouse, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), embryonal carcinoma cells (ECCs), and epiblast stem cells (EpiSCs). While all are pluripotent, these cell lines differ in terms of developmental origins, morphology, gene expression, and signaling, indicating that multiple pluripotent states exist. Whether and how the pluripotent state influences the cell line's developmental potential or the competence to respond to differentiation cues could help optimize directed differentiation protocols. To determine whether pluripotent stem cell lines differ in developmental potential, we compared the capacity of mouse ESCs, iPSCs, ECCs, and EpiSCs to form trophoblast. ESCs do not readily differentiate into trophoblast, but overexpression of the trophoblast-expressed transcription factor, CDX2, leads to efficient differentiation to trophoblast and to formation of trophoblast stem cells (TSCs) in the presence of fibroblast growth factor-4 (FGF4) and Heparin. Interestingly, we found that iPSCs and ECCs could both give rise to TSC-like cells following Cdx2 overexpression, suggesting that these cell lines are equivalent in developmental potential. By contrast, EpiSCs did not give rise to TSCs following Cdx2 overexpression, indicating that EpiSCs are no longer competent to respond to CDX2 by differentiating to trophoblast. In addition, we noted that culturing ESCs in conditions that promote naïve pluripotency improved the efficiency with which TSC-like cells could be derived. This work demonstrates that CDX2 efficiently induces trophoblast in more naïve than in primed pluripotent stem cells and that the pluripotent state can influence the developmental potential of stem cell lines.
Collapse
Affiliation(s)
- Stephanie Blij
- 1Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California
| | - Anthony Parenti
- 2Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Neeloufar Tabatabai-Yazdi
- 1Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California
| | - Amy Ralston
- 2Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
565
|
Soares MJ, Chakraborty D, Kubota K, Renaud SJ, Rumi MAK. Adaptive mechanisms controlling uterine spiral artery remodeling during the establishment of pregnancy. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2015; 58:247-59. [PMID: 25023691 DOI: 10.1387/ijdb.140083ms] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Implantation of the embryo into the uterus triggers the initiation of hemochorial placentation. The hemochorial placenta facilitates the acquisition of maternal resources required for embryo/fetal growth. Uterine spiral arteries form the nutrient supply line for the placenta and fetus. This vascular conduit undergoes gestation stage-specific remodeling directed by maternal natural killer cells and embryo-derived invasive trophoblast lineages. The placentation site, including remodeling of the uterine spiral arteries, is shaped by environmental challenges. In this review, we discuss the cellular participants controlling pregnancy-dependent uterine spiral artery remodeling and mechanisms responsible for their development and function.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.
| | | | | | | | | |
Collapse
|
566
|
A gp130-Src-YAP module links inflammation to epithelial regeneration. Nature 2015; 519:57-62. [PMID: 25731159 DOI: 10.1038/nature14228] [Citation(s) in RCA: 515] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 01/09/2015] [Indexed: 02/06/2023]
Abstract
Inflammation promotes regeneration of injured tissues through poorly understood mechanisms, some of which involve interleukin (IL)-6 family members, the expression of which is elevated in many diseases including inflammatory bowel diseases and colorectal cancer. Here we show in mice and human cells that gp130, a co-receptor for IL-6 cytokines, triggers activation of YAP and Notch, transcriptional regulators that control tissue growth and regeneration, independently of the gp130 effector STAT3. Through YAP and Notch, intestinal gp130 signalling stimulates epithelial cell proliferation, causes aberrant differentiation and confers resistance to mucosal erosion. gp130 associates with the related tyrosine kinases Src and Yes, which are activated on receptor engagement to phosphorylate YAP and induce its stabilization and nuclear translocation. This signalling module is strongly activated upon mucosal injury to promote healing and maintain barrier function.
Collapse
|
567
|
Abstract
Lessons learned from conserved vertebrate developmental pathways have catalyzed rapid advances in pluripotent stem cell differentiation towards therapeutically relevant cell types. The most highly conserved phases of development are associated with the early patterning of the body plan - the so-called phylotypic stage. Both prior to and after this stage there is much more divergence across species. Developmental differences between human and mouse at the blastocyst and early post-implantation stages might help explain the differences among the different stem cell lines derived from these embryos. A better understanding of these early stages of human development will aid our ability to generate and manipulate human stem cells and their derivatives.
Collapse
Affiliation(s)
- Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, Ontario, Canada M5G OA4 Department of Molecular Genetics, University of Toronto, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, Ontario, Canada M5G OA4
| |
Collapse
|
568
|
Abstract
The early mouse embryo is an excellent system to study how a small group of initially rounded cells start to change shape and establish the first forms of adhesion-based cell-cell interactions in mammals in vivo. In addition to its critical role in the structural integrity of the embryo, we discuss here how adhesion is important to regulate cell polarity and cell fate. Recent evidence suggests that adherens junctions participate in signaling pathways by localizing key proteins to subcellular microdomains. E-cadherin has been identified as the main player required for the establishment of adhesion but other mechanisms involving additional proteins or physical forces acting in the embryo may also contribute. Application of new technologies that enable high-resolution quantitative imaging of adhesion protein dynamics and measurements of biomechanical forces will provide a greater understanding of how adhesion patterns the early mammalian embryo.
Collapse
Affiliation(s)
- Melanie D White
- European Molecular Biology Laboratory (EMBL) Australia, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Nicolas Plachta
- European Molecular Biology Laboratory (EMBL) Australia, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
569
|
Affiliation(s)
- Fa-Xing Yu
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
- Children's Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China 200032
| | - Zhipeng Meng
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
| | - Steven W. Plouffe
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
| |
Collapse
|
570
|
Taher L, Pfeiffer MJ, Fuellen G. Bioinformatics approaches to single-blastomere transcriptomics. Mol Hum Reprod 2015; 21:115-125. [PMID: 25239944 DOI: 10.1093/molehr/gau083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
The totipotent zygote gives rise to cells with differing identities during mouse preimplantation development. Many studies have focused on analyzing the spatio-temporal dependencies during these lineage decision processes and much has been learnt by tracing transgenic marker gene expression up to the blastocyst stage and by analyzing the effects of genetic manipulations (knockout/ overexpression) on embryo development. However, until recently, it has not been possible to get broader overviews on the gene expression networks that distinguish one cell from the other within the same embryo. With the advent of whole genome amplification methodology and microfluidics-based quantitative RT-PCR it became possible to generate transcriptomes of single cells. Here we review the current state of the art of single-cell transcriptomics applied to mouse preimplantation embryo blastomeres and summarize findings made by pioneering studies in recent years. Furthermore we use the PluriNetWork and ExprEssence to investigate cell transitions based on published data.
Collapse
Affiliation(s)
- Leila Taher
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Martin J Pfeiffer
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
571
|
Abstract
Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are the major downstream effectors of the Hippo pathway, which regulates tissue homeostasis, organ size, regeneration and tumorigenesis. In this Progress article, we summarize the current understanding of the biological functions of YAP and TAZ, and how the regulation of these two proteins can be disrupted in cancer. We also highlight recent findings on their expanding role in cancer progression and describe the potential of these targets for therapeutic intervention.
Collapse
Affiliation(s)
- Toshiro Moroishi
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Carsten Gram Hansen
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
572
|
Abstract
Although hormonal regulation of ovarian follicle development has been extensively investigated, most studies concentrate on the development of early antral follicles to the preovulatory stage, leading to the successful use of exogenous FSH for infertility treatment. Accumulating data indicate that preantral follicles are under stringent regulation by FSH and local intraovarian factors, thus providing the possibility to develop new therapeutic approaches. Granulosa cell-derived C-type natriuretic factor not only suppresses the final maturation of oocytes to undergo germinal vesicle breakdown before ovulation but also promotes preantral and antral follicle growth. In addition, several oocyte- and granulosa cell-derived factors stimulate preantral follicle growth by acting through wingless, receptor tyrosine kinase, receptor serine kinase, and other signaling pathways. In contrast, the ovarian Hippo signaling pathway constrains follicle growth and disruption of Hippo signaling promotes the secretion of downstream CCN growth factors capable of promoting follicle growth. Although the exact hormonal factors involved in primordial follicle activation has yet to be elucidated, the protein kinase B (AKT) and mammalian target of rapamycin signaling pathways are important for the activation of dormant primordial follicles. Hippo signaling disruption after ovarian fragmentation, combined with treating ovarian fragments with phosphatase and tensin homolog (PTEN) inhibitors and phosphoinositide-3-kinase stimulators to augment AKT signaling, promote the growth of preantral follicles in patients with primary ovarian insufficiency, leading to a new infertility intervention for such patients. Elucidation of intraovarian mechanisms underlying early folliculogenesis may allow the development of novel therapeutic strategies for patients diagnosed with primary ovarian insufficiency, polycystic ovary syndrome, and poor ovarian response to FSH stimulation, as well as for infertile women of advanced reproductive age.
Collapse
Affiliation(s)
- Aaron J W Hsueh
- Program of Reproductive and Stem Cell Biology (A.J.W.H., Y.C.), Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305-5317; Department of Obstetrics and Gynecology (K.K.), St. Mariana University School of Medicine, Kawasaki, Kanagawa 216-8511, Japan; Department of Reproductive Medicine & Gynecology (B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | | | | | | |
Collapse
|
573
|
Zhou LQ, Dean J. Reprogramming the genome to totipotency in mouse embryos. Trends Cell Biol 2015; 25:82-91. [PMID: 25448353 PMCID: PMC4312727 DOI: 10.1016/j.tcb.2014.09.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/17/2014] [Accepted: 09/23/2014] [Indexed: 02/03/2023]
Abstract
Despite investigative interest, the artificial derivation of pluripotent stem cells remains inefficient and incomplete reprogramming hinders its potential as a reliable tool in regenerative medicine. By contrast, fusion of terminally differentiated gametes at fertilization activates efficient epigenetic reprogramming to ensure totipotency of early embryos. Understanding the epigenetic mechanisms required for the transition from the fertilized egg to the embryo can improve efforts to reprogram differentiated cells to pluripotent/totipotent cells for therapeutic use. We review recent discoveries that are providing insight into the molecular mechanisms required for epigenetic reprogramming to totipotency in vivo.
Collapse
Affiliation(s)
- Li-quan Zhou
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jurrien Dean
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
574
|
Liu S, Bou G, Sun R, Guo S, Xue B, Wei R, Cooney AJ, Liu Z. Sox2 is the faithful marker for pluripotency in pig: evidence from embryonic studies. Dev Dyn 2015; 244:619-27. [PMID: 25619399 DOI: 10.1002/dvdy.24248] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 12/25/2014] [Accepted: 12/26/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mammalian first lineage segregation generates trophectoderm (TE) and pluripotent inner cell mass (ICM), which provides an ideal model for studying the mechanisms of maintenance and loss of pluripotency. In mouse, the transcription factor OCT4 restricts to ICM and plays a key role in TE/ICM specification and pluripotent regulatory networks. However, in pig, OCT4 does not restrict to ICM cells, suggesting a different molecular basis in TE/ICM specification and pluripotent regulatory networks. RESULTS To explore molecular basis of porcine TE/ICM specification and pluripotent regulatory networks, we examined expression pattern of pluripotency factors, including SOX2, REX1, SALL4, ESG1, NANOG, TBX3, LIN28, KLF2, and KLF5, in porcine blastocysts. We found that SOX2 is a faithful pluripotent marker that anchored to the pluripotent cells including embryonic part cells, ICM cells and newly EPI cells along with developmental progress, whereas OCT4 expressed in almost all the cells at the same time. Consistently, analysis of spatiotemporal distribution of SOX2 and the TE marker CDX2 revealed an exclusive expression pattern in D6 blastocysts, whereas no correlation was observed between OCT4 and CDX2 at the same stage. CONCLUSIONS Our results provide a molecular basis in porcine embryonic patterning and a clue for further studying porcine pluripotent regulatory networks.
Collapse
Affiliation(s)
- Shichao Liu
- College of life science, Northeast Agricultural University of China, Harbin, China
| | | | | | | | | | | | | | | |
Collapse
|
575
|
Tajonar A, Maehr R, Hu G, Sneddon JB, Rivera-Feliciano J, Cohen DE, Elledge SJ, Melton DA. Brief report: VGLL4 is a novel regulator of survival in human embryonic stem cells. Stem Cells 2015; 31:2833-41. [PMID: 23765749 PMCID: PMC4617635 DOI: 10.1002/stem.1445] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 02/21/2013] [Accepted: 03/07/2013] [Indexed: 01/07/2023]
Abstract
Human embryonic stem cells (hESCs) are maintained in a self-renewing state by an interconnected network of mechanisms that sustain pluripotency, promote proliferation and survival, and prevent differentiation. We sought to find novel genes that could contribute to one or more of these processes using a gain-of-function screen of a large collection of human open reading frames. We identified Vestigial-like 4 (VGLL4), a cotranscriptional regulator with no previously described function in hESCs, as a positive regulator of survival in hESCs. Specifically, VGLL4 overexpression in hESCs significantly decreases cell death in response to dissociation stress. Additionally, VGLL4 overexpression enhances hESC colony formation from single cells. These effects may be attributable, in part, to a decreased activity of initiator and effector caspases observed in the context of VGLL4 overexpression. Additionally, we show an interaction between VGLL4 and the Rho/Rock pathway, previously implicated in hESC survival. This study introduces a novel gain-of-function approach for studying hESC maintenance and presents VGLL4 as a previously undescribed regulator of this process. Stem Cells 2013;31:2833-2841.
Collapse
Affiliation(s)
- Adriana Tajonar
- Department of Stem Cell and Regenerative Biology, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
576
|
Wang LH, Baker NE. Salvador-Warts-Hippo pathway in a developmental checkpoint monitoring helix-loop-helix proteins. Dev Cell 2015; 32:191-202. [PMID: 25579975 DOI: 10.1016/j.devcel.2014.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 10/16/2014] [Accepted: 12/01/2014] [Indexed: 12/19/2022]
Abstract
The E proteins and Id proteins are, respectively, the positive and negative heterodimer partners for the basic-helix-loop-helix protein family and as such contribute to a remarkably large number of cell-fate decisions. E proteins and Id proteins also function to inhibit or promote cell proliferation and cancer. Using a genetic modifier screen in Drosophila, we show that the Id protein Extramacrochaetae enables growth by suppressing activation of the Salvador-Warts-Hippo pathway of tumor suppressors, activation that requires transcriptional activation of the expanded gene by the E protein Daughterless. Daughterless protein binds to an intronic enhancer in the expanded gene, both activating the SWH pathway independently of the transmembrane protein Crumbs and bypassing the negative feedback regulation that targets the same expanded enhancer. Thus, the Salvador-Warts-Hippo pathway has a cell-autonomous function to prevent inappropriate differentiation due to transcription factor imbalance and monitors the intrinsic developmental status of progenitor cells, distinct from any responses to cell-cell interactions.
Collapse
Affiliation(s)
- Lan-Hsin Wang
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Nicholas E Baker
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
577
|
Kodaka M, Hata Y. The mammalian Hippo pathway: regulation and function of YAP1 and TAZ. Cell Mol Life Sci 2015; 72:285-306. [PMID: 25266986 PMCID: PMC11113917 DOI: 10.1007/s00018-014-1742-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/08/2014] [Accepted: 09/25/2014] [Indexed: 02/07/2023]
Abstract
The Hippo pathway was originally identified as the signaling that controls organ size in Drosophila, with the core architecture conserved in mammals. In the mammalian Hippo pathway, mammalian Ste20-like kinases (MST1/2) and large tumor suppressor kinases (LATS1/2) regulate transcriptional co-activators, Yes-associated protein (YAP1) and Transcriptional co-activator with a PDZ-binding motif (TAZ). The Hippo pathway was initially thought to be quite straightforward; however, the identification of additional components has revealed its inherent complexity. Regulation of YAP1 and TAZ is not always dependent on MST1/2 and LATS1/2. MST1/2 and LATS1/2 play various YAP1/TAZ-independent roles, while YAP1 and TAZ cross-talk with other signaling pathways. In this review we focus on YAP1 and TAZ and discuss their regulation, function, and the consequences of their dysregulation.
Collapse
Affiliation(s)
- Manami Kodaka
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8519 Japan
| | - Yutaka Hata
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8519 Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, 113-8519 Japan
| |
Collapse
|
578
|
Zhu C, Li L, Zhao B. The regulation and function of YAP transcription co-activator. Acta Biochim Biophys Sin (Shanghai) 2015; 47:16-28. [PMID: 25487920 DOI: 10.1093/abbs/gmu110] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Hippo pathway was initially identified in Drosophila by genetic mosaic screens for tumor suppressor genes. Researches indicated that the Hippo pathway is a key regulator of organ size and is conserved during evolution. Furthermore, studies of mouse models and clinical samples demonstrated the importance of Hippo pathway dysregulation in human cancer development. In addition, the Hippo pathway contributes to progenitor cell and stem cell self-renewal and is thus involved in tissue regeneration. In the Hippo pathway, MST1/2 kinases together with the adaptor protein SAV phosphorylate LATS1/2 kinases. Interaction with an adaptor protein MOB is also important for LATS1/2 activation. Activated LATS1/2 in turn phosphorylate and inhibit Yes-associated protein (YAP). YAP is a key downstream effector of the Hippo pathway, and is a transcriptional co-activator that mainly interacts with TEAD family transcription factors to promote gene expression. Alteration of gene expression by YAP leads to cell proliferation, apoptosis evasion, and also stem cell amplification. In this review, we mainly focus on YAP, discussing its regulation and mechanisms of action in the context of organ size control, tissue regeneration and tumorigenesis.
Collapse
Affiliation(s)
- Chu Zhu
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou 310058, China
| | - Li Li
- Institute of Aging Research, Hangzhou Normal University, Hangzhou 311121, China
| | - Bin Zhao
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
579
|
Zhou GX, Li XY, Zhang Q, Zhao K, Zhang CP, Xue CH, Yang K, Tian ZB. Effects of the hippo signaling pathway in human gastric cancer. Asian Pac J Cancer Prev 2014; 14:5199-205. [PMID: 24175801 DOI: 10.7314/apjcp.2013.14.9.5199] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND/AIM The Hippo signaling pathway is a newly discovered and conserved signaling cascade, which regulates organ size control by governing cell proliferation and apoptosis. This study aimed to investigate its effects in human gastric cancer. METHODS Tumor tissues (n=60), adjacent non-tumor tissues (n=60) and normal tissues (n=60) were obtained from the same patients with primary gastric cancer (GC). In addition, 70 samples of chronic atrophic gastritis (CAG) tissues were obtained from patients with intestinal metaplasia (IM) by endoscopic biopsy. Hippo signaling molecules, including Mst1, Lats1, YAP1, TAZ, TEAD1, Oct4 and CDX2, were determined by quantitative polymerase chain reaction (qPCR). Protein expression of Mst1, Lats1, YAP1, TEAD1 and CDX2 was assessed by immunohistochemistry and Western blotting. RESULTS Mst1, Lats1 and Oct4 mRNA expression showed an increasing tendency from GC tissues to normal gastric tissues, while the mRNA expression of YAP1, TAZ and TEAD1 was up-regulated (all P<0.01). Mst1 and Lats1 protein expression presented a similar trend with their mRNA expression. In addition, YAP1 and TEAD1 protein expression in GC was significantly higher than in the other groups (all P<0.01). CDX2 mRNA and protein expression in the CAG group were higher than in the other groups (all P<0.01). In GC, mRNA expression of Mst1, Lats1, Oct4, YAP1, TAZ, TEAD1 and CDX2 had a close correlation with lymphatic metastasis and tumor TNM stage (all P<0.01). Furthermore, protein expression of Mst1, Lats1 ,YAP1, TAZ, TEAD1 and CDX2 had a close correlation between each other (P<0.05). CONCLUSION The Hippo signaling pathway is involved in the development, progression and metastasis of human gastric cancer. Therefore, manipulation of Hippo signaling molecules may be a potential therapeutic strategy for gastric cancer.
Collapse
Affiliation(s)
- Guang-Xi Zhou
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University Medical College, Qingdao, China E-mail :
| | | | | | | | | | | | | | | |
Collapse
|
580
|
Imajo M, Ebisuya M, Nishida E. Dual role of YAP and TAZ in renewal of the intestinal epithelium. Nat Cell Biol 2014; 17:7-19. [DOI: 10.1038/ncb3084] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/13/2014] [Indexed: 02/07/2023]
|
581
|
Jedrusik A, Cox A, Wicher KB, Glover DM, Zernicka-Goetz M. Maternal-zygotic knockout reveals a critical role of Cdx2 in the morula to blastocyst transition. Dev Biol 2014; 398:147-52. [PMID: 25512302 PMCID: PMC4319684 DOI: 10.1016/j.ydbio.2014.12.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 12/06/2014] [Indexed: 12/01/2022]
Abstract
The first lineage segregation in the mouse embryo generates the inner cell mass (ICM), which gives rise to the pluripotent epiblast and therefore the future embryo, and the trophectoderm (TE), which will build the placenta. The TE lineage depends on the transcription factor Cdx2. However, when Cdx2 first starts to act remains unclear. Embryos with zygotic deletion of Cdx2 develop normally until the late blastocyst stage leading to the conclusion that Cdx2 is important for the maintenance but not specification of the TE. In contrast, down-regulation of Cdx2 transcripts from the early embryo stage results in defects in TE specification before the blastocyst stage. Here, to unambiguously address at which developmental stage Cdx2 becomes first required, we genetically deleted Cdx2 from the oocyte stage using a Zp3-Cre/loxP strategy. Careful assessment of a large cohort of Cdx2 maternal-zygotic null embryos, all individually filmed, examined and genotyped, reveals an earlier lethal phenotype than observed in Cdx2 zygotic null embryos that develop until the late blastocyst stage. The developmental failure of Cdx2 maternal-zygotic null embryos is associated with cell death and failure of TE specification, starting at the morula stage. These results indicate that Cdx2 is important for the correct specification of TE from the morula stage onwards and that both maternal and zygotic pools of Cdx2 are required for correct pre-implantation embryogenesis.
Collapse
Affiliation(s)
- Agnieszka Jedrusik
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3DY, UK
| | - Andy Cox
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3DY, UK
| | - Krzysztof B Wicher
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3DY, UK
| | - David M Glover
- Department of Genetics, University of Cambridge, Downing Site, Cambridge CB2 3EH, UK
| | - Magdalena Zernicka-Goetz
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3DY, UK
| |
Collapse
|
582
|
Rhee C, Lee BK, Beck S, Anjum A, Cook KR, Popowski M, Tucker HO, Kim J. Arid3a is essential to execution of the first cell fate decision via direct embryonic and extraembryonic transcriptional regulation. Genes Dev 2014; 28:2219-32. [PMID: 25319825 PMCID: PMC4201284 DOI: 10.1101/gad.247163.114] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Despite their origin from the inner cell mass, embryonic stem (ES) cells undergo differentiation to the trophectoderm (TE) lineage by repression of the ES cell master regulator Oct4 or activation of the TE master regulator Caudal-type homeobox 2 (Cdx2). In contrast to the in-depth studies of ES cell self-renewal and pluripotency, few TE-specific regulators have been identified, thereby limiting our understanding of mechanisms underlying the first cell fate decision. Here we show that up-regulation and nuclear entry of AT-rich interactive domain 3a (Arid3a) drives TE-like transcriptional programs in ES cells, maintains trophoblast stem (TS) cell self-renewal, and promotes further trophoblastic differentiation both upstream and independent of Cdx2. Accordingly, Arid3a(-/-) mouse post-implantation placental development is severely impaired, resulting in early embryonic death. We provide evidence that Arid3a directly activates TE-specific and trophoblast lineage-specific genes while directly repressing pluripotency genes via differential regulation of epigenetic acetylation or deacetylation. Our results identify Arid3a as a critical regulator of TE and placental development through execution of the commitment and differentiation phases of the first cell fate decision.
Collapse
Affiliation(s)
- Catherine Rhee
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology
| | - Bum-Kyu Lee
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology
| | - Samuel Beck
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology
| | | | | | - Melissa Popowski
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology
| | - Haley O Tucker
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology,
| | - Jonghwan Kim
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas 78712, USA
| |
Collapse
|
583
|
Piccolo S, Dupont S, Cordenonsi M. The biology of YAP/TAZ: hippo signaling and beyond. Physiol Rev 2014; 94:1287-312. [PMID: 25287865 DOI: 10.1152/physrev.00005.2014] [Citation(s) in RCA: 1292] [Impact Index Per Article: 117.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The transcriptional regulators YAP and TAZ are the focus of intense interest given their remarkable biological properties in development, tissue homeostasis and cancer. YAP and TAZ activity is key for the growth of whole organs, for amplification of tissue-specific progenitor cells during tissue renewal and regeneration, and for cell proliferation. In tumors, YAP/TAZ can reprogram cancer cells into cancer stem cells and incite tumor initiation, progression and metastasis. As such, YAP/TAZ are appealing therapeutic targets in cancer and regenerative medicine. Just like the function of YAP/TAZ offers a molecular entry point into the mysteries of tissue biology, their regulation by upstream cues is equally captivating. YAP/TAZ are well known for being the effectors of the Hippo signaling cascade, and mouse mutants in Hippo pathway components display remarkable phenotypes of organ overgrowth, enhanced stem cell content and reduced cellular differentiation. YAP/TAZ are primary sensors of the cell's physical nature, as defined by cell structure, shape and polarity. YAP/TAZ activation also reflects the cell "social" behavior, including cell adhesion and the mechanical signals that the cell receives from tissue architecture and surrounding extracellular matrix (ECM). At the same time, YAP/TAZ entertain relationships with morphogenetic signals, such as Wnt growth factors, and are also regulated by Rho, GPCRs and mevalonate metabolism. YAP/TAZ thus appear at the centerpiece of a signaling nexus by which cells take control of their behavior according to their own shape, spatial location and growth factor context.
Collapse
Affiliation(s)
- Stefano Piccolo
- Department of Molecular Medicine, University of Padua School of Medicine, Padua, Italy
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padua School of Medicine, Padua, Italy
| | | |
Collapse
|
584
|
Abstract
In mice, three pluripotent stem cell lines have been established from different stage of developing embryo, which are embryonic stem (ES) cell, post-implantation epiblast stem cell (EpiSC), and embryonic germ (EG) cell. ES cell and EG cell share many common features including factor requirement, colony morphology, and gene expression pattern. On the other hand, EpiSC needs different external signal inputs, exhibits flattened colony morphology, and a different set of gene expression patterns. In addition, the germ line competency of EpiSCs is still unclear. To distinguish the differences between them, they are defined by the words "naïve" and "primed" pluripotent cells, respectively. This article introduces how pluripotent stem cell lines are established in culture, and how much those cells in vitro are similar or relevant to their in vivo origin and the knowledge about transcription factors to support this state.
Collapse
|
585
|
Hayashi S, Ochi H, Ogino H, Kawasumi A, Kamei Y, Tamura K, Yokoyama H. Transcriptional regulators in the Hippo signaling pathway control organ growth in Xenopus tadpole tail regeneration. Dev Biol 2014; 396:31-41. [DOI: 10.1016/j.ydbio.2014.09.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/06/2014] [Accepted: 09/17/2014] [Indexed: 11/28/2022]
|
586
|
Abstract
Polarization of early embryos along cell contact patterns—referred to in this paper as radial polarization—provides a foundation for the initial cell fate decisions and morphogenetic movements of embryogenesis. Although polarity can be established through distinct upstream mechanisms in Caenorhabditis elegans, Xenopus laevis, and mouse embryos, in each species, it results in the restriction of PAR polarity proteins to contact-free surfaces of blastomeres. In turn, PAR proteins influence cell fates by affecting signaling pathways, such as Hippo and Wnt, and regulate morphogenetic movements by directing cytoskeletal asymmetries.
Collapse
Affiliation(s)
- Jeremy Nance
- Helen L. and Martin S. Kimmel Center for Biology and Medicine, the Skirball Institute of Biomolecular Medicine, and Department of Cell Biology, New York University School of Medicine, New York, NY 10016 Helen L. and Martin S. Kimmel Center for Biology and Medicine, the Skirball Institute of Biomolecular Medicine, and Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
587
|
Cambuli F, Murray A, Dean W, Dudzinska D, Krueger F, Andrews S, Senner CE, Cook SJ, Hemberger M. Epigenetic memory of the first cell fate decision prevents complete ES cell reprogramming into trophoblast. Nat Commun 2014; 5:5538. [PMID: 25423963 PMCID: PMC4263130 DOI: 10.1038/ncomms6538] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 10/10/2014] [Indexed: 12/17/2022] Open
Abstract
Embryonic (ES) and trophoblast (TS) stem cells reflect the first, irrevocable cell fate decision in development that is reinforced by distinct epigenetic lineage barriers. Nonetheless, ES cells can seemingly acquire TS-like characteristics upon manipulation of lineage-determining transcription factors or activation of the extracellular signal-regulated kinase 1/2 (Erk1/2) pathway. Here we have interrogated the progression of reprogramming in ES cell models with regulatable Oct4 and Cdx2 transgenes or conditional Erk1/2 activation. Although trans-differentiation into TS-like cells is initiated, lineage conversion remains incomplete in all models, underpinned by the failure to demethylate a small group of TS cell genes. Forced expression of these non-reprogrammed genes improves trans-differentiation efficiency, but still fails to confer a stable TS cell phenotype. Thus, even ES cells in ground-state pluripotency cannot fully overcome the boundaries that separate the first cell lineages but retain an epigenetic memory of their ES cell origin.
Collapse
Affiliation(s)
- Francesco Cambuli
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Alexander Murray
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Wendy Dean
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Dominika Dudzinska
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Felix Krueger
- Bioinformatics Group, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Simon Andrews
- Bioinformatics Group, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Claire E. Senner
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Simon J. Cook
- Signalling Programme, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Myriam Hemberger
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
- Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
588
|
Chen H, Guo J, Mishra SK, Robson P, Niranjan M, Zheng J. Single-cell transcriptional analysis to uncover regulatory circuits driving cell fate decisions in early mouse development. ACTA ACUST UNITED AC 2014; 31:1060-6. [PMID: 25416748 DOI: 10.1093/bioinformatics/btu777] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/17/2014] [Indexed: 11/12/2022]
Abstract
MOTIVATION Transcriptional regulatory networks controlling cell fate decisions in mammalian embryonic development remain elusive despite a long time of research. The recent emergence of single-cell RNA profiling technology raises hope for new discovery. Although experimental works have obtained intriguing insights into the mouse early development, a holistic and systematic view is still missing. Mathematical models of cell fates tend to be concept-based, not designed to learn from real data. To elucidate the regulatory mechanisms behind cell fate decisions, it is highly desirable to synthesize the data-driven and knowledge-driven modeling approaches. RESULTS We propose a novel method that integrates the structure of a cell lineage tree with transcriptional patterns from single-cell data. This method adopts probabilistic Boolean network (PBN) for network modeling, and genetic algorithm as search strategy. Guided by the 'directionality' of cell development along branches of the cell lineage tree, our method is able to accurately infer the regulatory circuits from single-cell gene expression data, in a holistic way. Applied on the single-cell transcriptional data of mouse preimplantation development, our algorithm outperforms conventional methods of network inference. Given the network topology, our method can also identify the operational interactions in the gene regulatory network (GRN), corresponding to specific cell fate determination. This is one of the first attempts to infer GRNs from single-cell transcriptional data, incorporating dynamics of cell development along a cell lineage tree. AVAILABILITY AND IMPLEMENTATION Implementation of our algorithm is available from the authors upon request. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Haifen Chen
- School of Computer Engineering, Nanyang Technological University, Singapore 639798, Singapore, Genome Institute of Singapore, Biopolis, Singapore 138672, Singapore and School of Electronics and Computer Science, University of Southampton, Highfield, Southampton SO17 1BJ, UK
| | - Jing Guo
- School of Computer Engineering, Nanyang Technological University, Singapore 639798, Singapore, Genome Institute of Singapore, Biopolis, Singapore 138672, Singapore and School of Electronics and Computer Science, University of Southampton, Highfield, Southampton SO17 1BJ, UK
| | - Shital K Mishra
- School of Computer Engineering, Nanyang Technological University, Singapore 639798, Singapore, Genome Institute of Singapore, Biopolis, Singapore 138672, Singapore and School of Electronics and Computer Science, University of Southampton, Highfield, Southampton SO17 1BJ, UK
| | - Paul Robson
- School of Computer Engineering, Nanyang Technological University, Singapore 639798, Singapore, Genome Institute of Singapore, Biopolis, Singapore 138672, Singapore and School of Electronics and Computer Science, University of Southampton, Highfield, Southampton SO17 1BJ, UK
| | - Mahesan Niranjan
- School of Computer Engineering, Nanyang Technological University, Singapore 639798, Singapore, Genome Institute of Singapore, Biopolis, Singapore 138672, Singapore and School of Electronics and Computer Science, University of Southampton, Highfield, Southampton SO17 1BJ, UK
| | - Jie Zheng
- School of Computer Engineering, Nanyang Technological University, Singapore 639798, Singapore, Genome Institute of Singapore, Biopolis, Singapore 138672, Singapore and School of Electronics and Computer Science, University of Southampton, Highfield, Southampton SO17 1BJ, UK School of Computer Engineering, Nanyang Technological University, Singapore 639798, Singapore, Genome Institute of Singapore, Biopolis, Singapore 138672, Singapore and School of Electronics and Computer Science, University of Southampton, Highfield, Southampton SO17 1BJ, UK
| |
Collapse
|
589
|
Milton CC, Grusche FA, Degoutin JL, Yu E, Dai Q, Lai EC, Harvey KF. The Hippo pathway regulates hematopoiesis in Drosophila melanogaster. Curr Biol 2014; 24:2673-80. [PMID: 25454587 PMCID: PMC4269548 DOI: 10.1016/j.cub.2014.10.031] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/26/2014] [Accepted: 10/13/2014] [Indexed: 12/13/2022]
Abstract
The Salvador-Warts-Hippo (Hippo) pathway is an evolutionarily conserved regulator of organ growth and cell fate. It performs these functions in epithelial and neural tissues of both insects and mammals, as well as in mammalian organs such as the liver and heart. Despite rapid advances in Hippo pathway research, a definitive role for this pathway in hematopoiesis has remained enigmatic. The hematopoietic compartments of Drosophila melanogaster and mammals possess several conserved features. D. melanogaster possess three types of hematopoietic cells that most closely resemble mammalian myeloid cells: plasmatocytes (macrophage-like cells), crystal cells (involved in wound healing), and lamellocytes (which encapsulate parasites). The proteins that control differentiation of these cells also control important blood lineage decisions in mammals. Here, we define the Hippo pathway as a key mediator of hematopoiesis by showing that it controls differentiation and proliferation of the two major types of D. melanogaster blood cells, plasmatocytes and crystal cells. In animals lacking the downstream Hippo pathway kinase Warts, lymph gland cells overproliferated, differentiated prematurely, and often adopted a mixed lineage fate. The Hippo pathway regulated crystal cell numbers by both cell-autonomous and non-cell-autonomous mechanisms. Yorkie and its partner transcription factor Scalloped were found to regulate transcription of the Runx family transcription factor Lozenge, which is a key regulator of crystal cell fate. Further, Yorkie or Scalloped hyperactivation induced ectopic crystal cells in a non-cell-autonomous and Notch-pathway-dependent fashion.
Collapse
Affiliation(s)
- Claire C Milton
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Felix A Grusche
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Joffrey L Degoutin
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Eefang Yu
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Qi Dai
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Eric C Lai
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Kieran F Harvey
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
590
|
Yao F, Liu H, Li Z, Zhong C, Fang W. Down-regulation of LATS2 in non-small cell lung cancer promoted the growth and motility of cancer cells. Tumour Biol 2014; 36:2049-57. [DOI: 10.1007/s13277-014-2812-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/04/2014] [Indexed: 12/25/2022] Open
|
591
|
Wicklow E, Blij S, Frum T, Hirate Y, Lang RA, Sasaki H, Ralston A. HIPPO pathway members restrict SOX2 to the inner cell mass where it promotes ICM fates in the mouse blastocyst. PLoS Genet 2014; 10:e1004618. [PMID: 25340657 PMCID: PMC4207610 DOI: 10.1371/journal.pgen.1004618] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 07/21/2014] [Indexed: 12/15/2022] Open
Abstract
Pluripotent epiblast (EPI) cells, present in the inner cell mass (ICM) of the mouse blastocyst, are progenitors of both embryonic stem (ES) cells and the fetus. Discovering how pluripotency genes regulate cell fate decisions in the blastocyst provides a valuable way to understand how pluripotency is normally established. EPI cells are specified by two consecutive cell fate decisions. The first decision segregates ICM from trophectoderm (TE), an extraembryonic cell type. The second decision subdivides ICM into EPI and primitive endoderm (PE), another extraembryonic cell type. Here, we investigate the roles and regulation of the pluripotency gene Sox2 during blastocyst formation. First, we investigate the regulation of Sox2 patterning and show that SOX2 is restricted to ICM progenitors prior to blastocyst formation by members of the HIPPO pathway, independent of CDX2, the TE transcription factor that restricts Oct4 and Nanog to the ICM. Second, we investigate the requirement for Sox2 in cell fate specification during blastocyst formation. We show that neither maternal (M) nor zygotic (Z) Sox2 is required for blastocyst formation, nor for initial expression of the pluripotency genes Oct4 or Nanog in the ICM. Rather, Z Sox2 initially promotes development of the primitive endoderm (PE) non cell-autonomously via FGF4, and then later maintains expression of pluripotency genes in the ICM. The significance of these observations is that 1) ICM and TE genes are spatially patterned in parallel prior to blastocyst formation and 2) both the roles and regulation of Sox2 in the blastocyst are unique compared to other pluripotency factors such as Oct4 or Nanog. Pluripotent stem cells can give rise to any cell type in the body, making them an attractive tool for regenerative medicine. Pluripotent stem cells can be derived from the mammalian embryo at the blastocyst stage or they can be created from mature adult cells by reprogramming. During reprogramming, SOX2 helps establish pluripotency, but it is not clear how SOX2 establishes pluripotency in the blastocyst. We evaluated where SOX2 is present, how SOX2 is regulated, and where SOX2 is active during blastocyst formation. Our data show that the roles and the regulation of SOX2 are unique compared to other pluripotency/reprogramming factors, such as OCT4 and NANOG. SOX2 marks pluripotent cells earlier than do other factors, but does not regulate pluripotency until several days later. Rather, the earlier role of SOX2 is to help establish the yolk sac lineage, which is essential for gestation.
Collapse
Affiliation(s)
- Eryn Wicklow
- Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Stephanie Blij
- Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Tristan Frum
- Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Yoshikazu Hirate
- Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Richard A. Lang
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Hiroshi Sasaki
- Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Amy Ralston
- Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
- * E-mail:
| |
Collapse
|
592
|
Gumbiner BM, Kim NG. The Hippo-YAP signaling pathway and contact inhibition of growth. J Cell Sci 2014; 127:709-17. [PMID: 24532814 DOI: 10.1242/jcs.140103] [Citation(s) in RCA: 268] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Hippo-YAP pathway mediates the control of cell proliferation by contact inhibition as well as other attributes of the physical state of cells in tissues. Several mechanisms sense the spatial and physical organization of cells, and function through distinct upstream modules to stimulate Hippo-YAP signaling: adherens junction or cadherin-catenin complexes, epithelial polarity and tight junction complexes, the FAT-Dachsous morphogen pathway, as well as cell shape, actomyosin or mechanotransduction. Soluble extracellular factors also regulate Hippo pathway signaling, often inhibiting its activity. Indeed, the Hippo pathway mediates a reciprocal relationship between contact inhibition and mitogenic signaling. As a result, cells at the edges of a colony, a wound in a tissue or a tumor are more sensitive to ambient levels of growth factors and more likely to proliferate, migrate or differentiate through a YAP and/or TAZ-dependent process. Thus, the Hippo-YAP pathway senses and responds to the physical organization of cells in tissues and coordinates these physical cues with classic growth-factor-mediated signaling pathways. This Commentary is focused on the biological significance of Hippo-YAP signaling and how upstream regulatory modules of the pathway interact to produce biological outcomes.
Collapse
Affiliation(s)
- Barry M Gumbiner
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | |
Collapse
|
593
|
Goissis MD, Cibelli JB. Functional characterization of CDX2 during bovine preimplantation development in vitro. Mol Reprod Dev 2014; 81:962-70. [PMID: 25251051 DOI: 10.1002/mrd.22415] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/20/2014] [Indexed: 01/26/2023]
Abstract
Placental defects are common in bovine embryos produced using assisted reproductive techniques. A proper understanding of the events leading to inner cell mass (ICM) and trophectoderm (TE) specification could help identify the origins of such developmental failures. We focused on caudal-type homeobox transcription factor 2 (CDX2) since it has a specific role during TE differentiation in mouse embryos. Of all the preimplantation stages analyzed, CDX2 protein was present only at the blastocyst stage. To further understand the roles of CDX2 during bovine development, we depleted CDX2 mRNA; despite a significant loss of detectable protein, embryos were able to form blastocysts at the same rate as controls. Embryos lacking CDX2 did not show abnormalities in the number of TE, ICM, or total cells in the blastocyst. Expression of the developmentally important genes SOX2, POU5F1, and NANOG, or TE markers such as IFN-T and KRT18 were not affected by the reduction in CDX2 levels, nor was the localization of SOX2 and POU5F1 protein. Using a functional barrier assay, we observed that the TE epithelial layer of embryos lacking CDX2 had lost its integrity. Our results thus indicate that CDX2 is not required for TE formation during bovine development; nevertheless, it is necessary for maintaining TE integrity.
Collapse
Affiliation(s)
- Marcelo D Goissis
- Department of Animal Science, Michigan State University, East Lansing, Michigan; Capes Foundation, Ministry of Education, Brasília, Brazil
| | | |
Collapse
|
594
|
Developmental pathways hijacked by osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:93-118. [PMID: 24924170 DOI: 10.1007/978-3-319-04843-7_5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cancer of any type often can be described by an arrest, alteration or disruption in the normal development of a tissue or organ, and understanding of the normal counterpart's development can aid in understanding the malignant state. This is certainly true for osteosarcoma and the normal developmental pathways that guide osteoblast development that are changed in the genesis of osteogenic sarcoma. A carefully regulated crescendo-decrescendo expression of RUNX2 accompanies the transition from mesenchymal stem cell to immature osteoblast to mature osteoblast. This pivotal role is controlled by several pathways, including bone morphogenic protein (BMP), Wnt/β-catenin, fibroblast growth factor (FGF), and protein kinase C (PKC). The HIPPO pathway and its downstream target YAP help to regulate proliferation of immature osteoblasts and their maturation into non-proliferating mature osteoblasts. This pathway also helps regulate expression of the mature osteoblast protein osteocalcin. YAP also regulates expression of MT1-MMP, a membrane-bound matrix metalloprotease responsible for remodeling the extracellular matrix surrounding the osteoblasts. YAP, in turn, can be regulated by the ERBB family protein Her-4. Osteosarcoma may be thought of as a cell held at the immature osteoblast stage, retaining some of the characteristics of that developmental stage. Disruptions of several of these pathways have been described in osteosarcoma, including BMP, Wnt/b-catenin, RUNX2, HIPPO/YAP, and Her-4. Further, PKC can be activated by several receptor tyrosine kinases implicated in osteosarcoma, including the ERBB family (EGFR, Her-2 and Her-4 in osteosarcoma), IGF1R, FGF, and others. Understanding these functions may aid in the understanding the mechanisms underpinning clinical observations in osteosarcoma, including both the lytic and blastic phenotypes of tumors, the invasiveness of the disease, and the tendency for treated tumors to ossify rather than shrink. Through a better understanding of the relationship between normal osteoblast development and osteosarcoma, we may gain insights into novel therapeutic avenues and improved outcomes.
Collapse
|
595
|
Knott JG, Paul S. Transcriptional regulators of the trophoblast lineage in mammals with hemochorial placentation. Reproduction 2014; 148:R121-36. [PMID: 25190503 DOI: 10.1530/rep-14-0072] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mammalian reproduction is critically dependent on the trophoblast cell lineage, which assures proper establishment of maternal-fetal interactions during pregnancy. Specification of trophoblast cell lineage begins with the development of the trophectoderm (TE) in preimplantation embryos. Subsequently, other trophoblast cell types arise with the progression of pregnancy. Studies with transgenic animal models as well as trophoblast stem/progenitor cells have implicated distinct transcriptional and epigenetic regulators in trophoblast lineage development. This review focuses on our current understanding of transcriptional and epigenetic mechanisms regulating specification, determination, maintenance and differentiation of trophoblast cells.
Collapse
Affiliation(s)
- Jason G Knott
- Developmental Epigenetics LaboratoryDepartment of Animal Science, Michigan State University, East Lansing, Michigan 48824, USADepartment of Pathology and Laboratory MedicineInstitute of Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Soumen Paul
- Developmental Epigenetics LaboratoryDepartment of Animal Science, Michigan State University, East Lansing, Michigan 48824, USADepartment of Pathology and Laboratory MedicineInstitute of Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| |
Collapse
|
596
|
Artus J, Chazaud C. A close look at the mammalian blastocyst: epiblast and primitive endoderm formation. Cell Mol Life Sci 2014; 71:3327-38. [PMID: 24794628 PMCID: PMC11113690 DOI: 10.1007/s00018-014-1630-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 04/14/2014] [Accepted: 04/15/2014] [Indexed: 10/25/2022]
Abstract
During early development, the mammalian embryo undergoes a series of profound changes that lead to the formation of two extraembryonic tissues--the trophectoderm and the primitive endoderm. These tissues encapsulate the pluripotent epiblast at the time of implantation. The current model proposes that the formation of these lineages results from two consecutive binary cell fate decisions. The first controls the formation of the trophectoderm and the inner cell mass, and the second controls the formation of the primitive endoderm and the epiblast within the inner cell mass. While early mammalian embryos develop with extensive plasticity, the embryonic pattern prior to implantation is remarkably reproducible. Here, we review the molecular mechanisms driving the cell fate decision between primitive endoderm and epiblast in the mouse embryo and integrate data from recent studies into the current model of the molecular network regulating the segregation between these lineages and their subsequent differentiation.
Collapse
Affiliation(s)
- Jérôme Artus
- Institut Pasteur, Mouse Functional Genetics, CNRS URA2578, 75015 Paris, France
| | - Claire Chazaud
- Clermont Université, Laboratoire GReD, Université d’Auvergne, BP 10448, 63000 Clermont-Ferrand, France
- Inserm, UMR1103, 63001 Clermont-Ferrand, France
- CNRS, UMR6293, 63001 Clermont-Ferrand, France
| |
Collapse
|
597
|
Sozen B, Can A, Demir N. Cell fate regulation during preimplantation development: a view of adhesion-linked molecular interactions. Dev Biol 2014; 395:73-83. [PMID: 25176042 DOI: 10.1016/j.ydbio.2014.08.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/20/2014] [Accepted: 08/21/2014] [Indexed: 12/01/2022]
Abstract
In the developmental process of the early mammalian embryo, it is crucial to understand how the identical cells in the early embryo later develop different fates. Along with existing models, many recently discovered molecular, cellular and developmental factors play roles in cell position, cell polarity and transcriptional networks in cell fate regulation during preimplantation. A structuring process known as compaction provides the "start signal" for cells to differentiate and orchestrates the developmental cascade. The proper intercellular junctional complexes assembled between blastomeres act as a conducting mechanism governing cellular diversification. Here, we provide an overview of the diversification process during preimplantation development as it relates to intercellular junctional complexes. We also evaluate transcriptional differences between embryonic lineages according to cell- cell adhesion and the contributions of adhesion to lineage commitment. These series of processes indicate that proper cell fate specification in the early mammalian embryo depends on junctional interactions and communication, which play essential roles during early morphogenesis.
Collapse
Affiliation(s)
- Berna Sozen
- Department of Histology and Embryology, School of Medicine, Akdeniz University, Campus, 07070 Antalya, Turkey
| | - Alp Can
- Department of Histology and Embryology, School of Medicine, Ankara University, Sihhiye, Ankara 06100, Turkey
| | - Necdet Demir
- Department of Histology and Embryology, School of Medicine, Akdeniz University, Campus, 07070 Antalya, Turkey.
| |
Collapse
|
598
|
Gao Y, Zhang W, Han X, Li F, Wang X, Wang R, Fang Z, Tong X, Yao S, Li F, Feng Y, Sun Y, Hou Y, Yang Z, Guan K, Chen H, Zhang L, Ji H. YAP inhibits squamous transdifferentiation of Lkb1-deficient lung adenocarcinoma through ZEB2-dependent DNp63 repression. Nat Commun 2014; 5:4629. [DOI: 10.1038/ncomms5629] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 07/08/2014] [Indexed: 02/07/2023] Open
|
599
|
Rayon T, Menchero S, Nieto A, Xenopoulos P, Crespo M, Cockburn K, Cañon S, Sasaki H, Hadjantonakis AK, de la Pompa JL, Rossant J, Manzanares M. Notch and hippo converge on Cdx2 to specify the trophectoderm lineage in the mouse blastocyst. Dev Cell 2014; 30:410-22. [PMID: 25127056 DOI: 10.1016/j.devcel.2014.06.019] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 05/12/2014] [Accepted: 06/23/2014] [Indexed: 11/15/2022]
Abstract
The first lineage choice in mammalian embryogenesis is that between the trophectoderm, which gives rise to the trophoblast of the placenta, and the inner cell mass, from which is derived the embryo proper and the yolk sac. The establishment of these lineages is preceded by the inside-versus-outside positioning of cells in the early embryo and stochastic expression of key transcription factors, which is then resolved into lineage-restricted expression. The regulatory inputs that drive this restriction and how they relate to cell position are largely unknown. Here, we show an unsuspected role of Notch signaling in regulating trophectoderm-specific expression of Cdx2 in cooperation with TEAD4. Notch activity is restricted to outer cells and is able to influence positional allocation of blastomeres, mediating preferential localization to the trophectoderm. Our results show that multiple signaling inputs at preimplantation stages specify the first embryonic lineages.
Collapse
Affiliation(s)
- Teresa Rayon
- Stem Cell Biology Program, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Sergio Menchero
- Stem Cell Biology Program, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Andres Nieto
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | | | - Miguel Crespo
- Stem Cell Biology Program, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Katie Cockburn
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Susana Cañon
- Stem Cell Biology Program, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Hiroshi Sasaki
- Institute of Molecular Embryology and Genetics, Department of Cell Fate Control, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | | | - Jose Luis de la Pompa
- Cardiovascular Developmental Biology Program, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Miguel Manzanares
- Stem Cell Biology Program, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| |
Collapse
|
600
|
Watanabe T, Biggins JS, Tannan NB, Srinivas S. Limited predictive value of blastomere angle of division in trophectoderm and inner cell mass specification. Development 2014; 141:2279-88. [PMID: 24866117 PMCID: PMC4034423 DOI: 10.1242/dev.103267] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The formation of trophectoderm (TE) and pluripotent inner cell mass (ICM) is one of the earliest events during mammalian embryogenesis. It is believed that the orientation of division of polarised blastomeres in the 8- and 16-cell stage embryo determines the fate of daughter cells, based on how asymmetrically distributed lineage determinants are segregated. To investigate the relationship between angle of division and subsequent fate in unperturbed embryos, we constructed cellular resolution digital representations of the development of mouse embryos from the morula to early blastocyst stage, based on 4D confocal image volumes. We find that at the 16-cell stage, very few inside cells are initially produced as a result of cell division, but that the number increases due to cell movement. Contrary to expectations, outside cells at the 16-cell stage represent a heterogeneous population, with some fated to contributing exclusively to the TE and others capable of contributing to both the TE and ICM. Our data support the view that factors other than the angle of division, such as the position of a blastomere, play a major role in the specification of TE and ICM.
Collapse
Affiliation(s)
- Tomoko Watanabe
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - John S Biggins
- Cavendish Laboratory, University of Cambridge, Cambridge CB3 0HE, UK
| | - Neeta Bala Tannan
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Shankar Srinivas
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| |
Collapse
|