1
|
Hammer A, Diakonova M. Prolactin-induced tyrosyl phosphorylation of PAK1 facilitates epithelial-mesenchymal transition. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001136. [PMID: 38660565 PMCID: PMC11040397 DOI: 10.17912/micropub.biology.001136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024]
Abstract
PAK1 and prolactin (PRL) regulate breast cancer. Prolactin-activated JAK2 tyrosyl phosphorylates PAK1 (pTyr-PAK1). We demonstrate here that pTyr-PAK1 regulates epithelial-mesenchymal transition (EMT) in breast cancer cells. PRL treatment of T47D PAK1 WT cells leads to downregulation of E-cadherin surface expression and "ectodomain shedding" (extracellular cleavage of E-cadherin). pTyr-PAK1 increases mRNA levels of Snail, Slug, and Twist2, transcriptional factors implicated in E-cadherin repression. pTyr-PAK1 also significantly increases PRL-dependent Slug activity leading to expression of vimentin, a hallmark of EMT. Thus, our current data on pTyr-PAK1 regulation of EMT bring insight into the role of PAK1 and PRL in human breast cancer.
Collapse
Affiliation(s)
- Alan Hammer
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States
| | - Maria Diakonova
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, United States
| |
Collapse
|
2
|
Windmöller BA, Höving AL, Knabbe C, Greiner JFW. Inter- and Intrapopulational Heterogeneity of Characteristic Markers in Adult Human Neural Crest-derived Stem Cells. Stem Cell Rev Rep 2021; 18:1510-1520. [PMID: 34748196 PMCID: PMC9033708 DOI: 10.1007/s12015-021-10277-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 11/24/2022]
Abstract
Adult human neural crest-derived stem cells (NCSCs) are found in a variety of adult tissues and show an extraordinary broad developmental potential. Despite their great differentiation capacity, increasing evidence suggest a remaining niche-dependent variability between different NCSC-populations regarding their differentiation behavior and expression signatures. In the present study, we extended the view on heterogeneity of NCSCs by identifying heterogeneous expression levels and protein amounts of characteristic markers even between NCSCs from the same niche of origin. In particular, populations of neural crest-derived inferior turbinate stem cells (ITSCs) isolated from different individuals showed significant variations in characteristic NCSC marker proteins Nestin, S100 and Slug in a donor-dependent manner. Notably, increased nuclear protein amounts of Slug were accompanied by a significantly elevated level of nuclear NF-κB-p65 protein, suggesting an NF-κB-dependent regulation of NCSC-makers. In addition to this interpopulational genetic heterogeneity of ITSC-populations from different donors, single ITSCs also revealed a strong heterogeneity regarding the protein amounts of Nestin, S100, Slug and NF-κB-p65 even within the same clonal culture. Our present findings therefor strongly suggest ITSC-heterogeneity to be at least partly based on an interpopulational genetic heterogeneity dependent on the donor accompanied by a stochastic intrapopulational heterogeneity between single cells. We propose this stochastic intrapopulational heterogeneity to occur in addition to the already described genetic variability between clonal NCSC-cultures and the niche-dependent plasticity of NCSCs. Our observations offer a novel perspective on NCSC-heterogeneity, which may build the basis to understand heterogeneous NCSC-behavior.
Collapse
Affiliation(s)
- Beatrice A Windmöller
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany.,Forschungsverbund BioMedizin Bielefeld FBMB e.V, Bielefeld, Germany.,Department of Cellular Neurophysiology, Faculty of Medicine, University of Bielefeld, Bielefeld, Germany
| | - Anna L Höving
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany.,Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545, Bad Oeynhausen, Germany
| | - Cornelius Knabbe
- Forschungsverbund BioMedizin Bielefeld FBMB e.V, Bielefeld, Germany.,Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545, Bad Oeynhausen, Germany
| | - Johannes F W Greiner
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany. .,Forschungsverbund BioMedizin Bielefeld FBMB e.V, Bielefeld, Germany.
| |
Collapse
|
3
|
Höving AL, Windmöller BA, Knabbe C, Kaltschmidt B, Kaltschmidt C, Greiner JFW. Between Fate Choice and Self-Renewal-Heterogeneity of Adult Neural Crest-Derived Stem Cells. Front Cell Dev Biol 2021; 9:662754. [PMID: 33898464 PMCID: PMC8060484 DOI: 10.3389/fcell.2021.662754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
Stem cells of the neural crest (NC) vitally participate to embryonic development, but also remain in distinct niches as quiescent neural crest-derived stem cell (NCSC) pools into adulthood. Although NCSC-populations share a high capacity for self-renewal and differentiation resulting in promising preclinical applications within the last two decades, inter- and intrapopulational differences exist in terms of their expression signatures and regenerative capability. Differentiation and self-renewal of stem cells in developmental and regenerative contexts are partially regulated by the niche or culture condition and further influenced by single cell decision processes, making cell-to-cell variation and heterogeneity critical for understanding adult stem cell populations. The present review summarizes current knowledge of the cellular heterogeneity within NCSC-populations located in distinct craniofacial and trunk niches including the nasal cavity, olfactory bulb, oral tissues or skin. We shed light on the impact of intrapopulational heterogeneity on fate specifications and plasticity of NCSCs in their niches in vivo as well as during in vitro culture. We further discuss underlying molecular regulators determining fate specifications of NCSCs, suggesting a regulatory network including NF-κB and NC-related transcription factors like SLUG and SOX9 accompanied by Wnt- and MAPK-signaling to orchestrate NCSC stemness and differentiation. In summary, adult NCSCs show a broad heterogeneity on the level of the donor and the donors' sex, the cell population and the single stem cell directly impacting their differentiation capability and fate choices in vivo and in vitro. The findings discussed here emphasize heterogeneity of NCSCs as a crucial parameter for understanding their role in tissue homeostasis and regeneration and for improving their applicability in regenerative medicine.
Collapse
Affiliation(s)
- Anna L. Höving
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre North Rhine-Westphalia (NRW), Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Beatrice A. Windmöller
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| | - Cornelius Knabbe
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre North Rhine-Westphalia (NRW), Ruhr University Bochum, Bad Oeynhausen, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| | - Johannes F. W. Greiner
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| |
Collapse
|
4
|
Klymkowsky MW. Filaments and phenotypes: cellular roles and orphan effects associated with mutations in cytoplasmic intermediate filament proteins. F1000Res 2019; 8. [PMID: 31602295 PMCID: PMC6774051 DOI: 10.12688/f1000research.19950.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2019] [Indexed: 12/11/2022] Open
Abstract
Cytoplasmic intermediate filaments (IFs) surround the nucleus and are often anchored at membrane sites to form effectively transcellular networks. Mutations in IF proteins (IFps) have revealed mechanical roles in epidermis, muscle, liver, and neurons. At the same time, there have been phenotypic surprises, illustrated by the ability to generate viable and fertile mice null for a number of IFp-encoding genes, including vimentin. Yet in humans, the vimentin ( VIM) gene displays a high probability of intolerance to loss-of-function mutations, indicating an essential role. A number of subtle and not so subtle IF-associated phenotypes have been identified, often linked to mechanical or metabolic stresses, some of which have been found to be ameliorated by the over-expression of molecular chaperones, suggesting that such phenotypes arise from what might be termed "orphan" effects as opposed to the absence of the IF network per se, an idea originally suggested by Toivola et al. and Pekny and Lane.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Molecular, Cellular & Developmental Biology, University of Colorado, Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
5
|
Li J, Perfetto M, Materna C, Li R, Thi Tran H, Vleminckx K, Duncan MK, Wei S. A new transgenic reporter line reveals Wnt-dependent Snai2 re-expression and cranial neural crest differentiation in Xenopus. Sci Rep 2019; 9:11191. [PMID: 31371771 PMCID: PMC6672020 DOI: 10.1038/s41598-019-47665-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
During vertebrate embryogenesis, the cranial neural crest (CNC) forms at the neural plate border and subsequently migrates and differentiates into many types of cells. The transcription factor Snai2, which is induced by canonical Wnt signaling to be expressed in the early CNC, is pivotal for CNC induction and migration in Xenopus. However, snai2 expression is silenced during CNC migration, and its roles at later developmental stages remain unclear. We generated a transgenic X. tropicalis line that expresses enhanced green fluorescent protein (eGFP) driven by the snai2 promoter/enhancer, and observed eGFP expression not only in the pre-migratory and migrating CNC, but also the differentiating CNC. This transgenic line can be used directly to detect deficiencies in CNC development at various stages, including subtle perturbation of CNC differentiation. In situ hybridization and immunohistochemistry confirm that Snai2 is re-expressed in the differentiating CNC. Using a separate transgenic Wnt reporter line, we show that canonical Wnt signaling is also active in the differentiating CNC. Blocking Wnt signaling shortly after CNC migration causes reduced snai2 expression and impaired differentiation of CNC-derived head cartilage structures. These results suggest that Wnt signaling is required for snai2 re-expression and CNC differentiation.
Collapse
Affiliation(s)
- Jiejing Li
- Department of Biology, West Virginia University, Morgantown, WV, 26506, USA.,Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming, 650032, China
| | - Mark Perfetto
- Department of Biology, West Virginia University, Morgantown, WV, 26506, USA.,Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Christopher Materna
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Rebecca Li
- Brown University, Providence, RI, 02912, USA
| | - Hong Thi Tran
- Department for Molecular Biomedical Research and Center for Medical Genetics, Ghent University, B-9052, Ghent, Belgium
| | - Kris Vleminckx
- Department for Molecular Biomedical Research and Center for Medical Genetics, Ghent University, B-9052, Ghent, Belgium
| | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
6
|
Rashid D, Puettmann P, Roy E, Bradley RS. Neural crest development in Xenopus requires Protocadherin 7 at the lateral neural crest border. Mech Dev 2018; 149:41-52. [PMID: 29366801 PMCID: PMC5820198 DOI: 10.1016/j.mod.2018.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/04/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022]
Abstract
In vertebrates, the neural crest is a unique population of pluripotent cells whose development is dependent on signaling from neighboring tissues. Cadherin family members, including protocadherins, are emerging as major players in neural crest development, largely through their roles in cell adhesion and sorting in embryonic tissues. Here, we show that Protocadherin 7 (Pcdh7), previously shown to function in sensorial layer integrity and neural tube closure in Xenopus, is also involved in neural crest specification and survival. Pcdh7 expression partly overlaps the neural crest domain at the lateral neural crest border. Pcdh7 knockdown in embryos does not alter neural crest induction; however, neural crest specification markers, including Snail2 and Sox9, are lost, due to apoptosis of the neural crest starting after stage 13. Pcdh7 knockdown also results in downregulation of Wnt11b; both of which are co-expressed in the sensorial layer lateral to the neural crest, suggestive of a role for Wnt11b in the neural crest apoptosis. Confirming this role, apoptosis, Snail2 expression and the developmental fate of the neural crest can be partially rescued by ectopic expression of Wnt11b. These results indicate that Pcdh7 plays an important role in maintaining the sensorial layer at the lateral neural crest border, which is necessary for the secretion of survival factors, including Wnt11b.
Collapse
Affiliation(s)
- Dana Rashid
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Paul Puettmann
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Ethan Roy
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Roger S. Bradley
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| |
Collapse
|
7
|
RCP induces Slug expression and cancer cell invasion by stabilizing β1 integrin. Oncogene 2016; 36:1102-1111. [PMID: 27524413 DOI: 10.1038/onc.2016.277] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 06/17/2016] [Accepted: 06/28/2016] [Indexed: 12/19/2022]
Abstract
Rab coupling protein (RCP)-induced tumor cell migration has been implicated in tumor pathophysiology and patient outcomes. In the present study, we demonstrate that RCP stabilizes β1 integrin leading to increased β1 integrin levels and activation of a signaling cascade culminating in Slug induction, epithelial-to-mesenchymal transition and increased invasion. Ectopic expression of RCP induced Slug expression. Silencing β1 integrin efficiently inhibited RCP-induced Slug expression and subsequent cancer cell invasion. Conversely, ectopic expression of β1 integrin was sufficient to induce Slug expression. Pharmacological inhibition of integrin linked kinase (ILK), EGFR and NF-κB, as well as transfection of a dominant-negative mutant of Ras (RasN17), significantly inhibited RCP-induced Slug expression and cancer cell invasion. Strikingly, ectopic expression of RCP was sufficient to enhance metastasis of ovarian cancer cells to the lung. Collectively, we demonstrate a mechanism by which RCP promotes cancer cell aggressiveness through sequential β1 integrin stabilization, activation of an ILK/EGFR/Ras/NF-κB signaling cascade and subsequent Slug expression.
Collapse
|
8
|
Macrì S, Simula L, Pellarin I, Pegoraro S, Onorati M, Sgarra R, Manfioletti G, Vignali R. Hmga2 is required for neural crest cell specification in Xenopus laevis. Dev Biol 2016; 411:25-37. [PMID: 26806704 DOI: 10.1016/j.ydbio.2016.01.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 12/23/2015] [Accepted: 01/20/2016] [Indexed: 12/12/2022]
Abstract
HMGA proteins are small nuclear proteins that bind DNA by conserved AT-hook motifs, modify chromatin architecture and assist in gene expression. Two HMGAs (HMGA1 and HMGA2), encoded by distinct genes, exist in mammals and are highly expressed during embryogenesis or reactivated in tumour progression. We here addressed the in vivo role of Xenopus hmga2 in the neural crest cells (NCCs). We show that hmga2 is required for normal NCC specification and development. hmga2 knockdown leads to severe disruption of major skeletal derivatives of anterior NCCs. We show that, within the NCC genetic network, hmga2 acts downstream of msx1, and is required for msx1, pax3 and snail2 activities, thus participating at different levels of the network. Because of hmga2 early effects in NCC specification, the subsequent epithelial-mesenchymal transition (EMT) and migration of NCCs towards the branchial pouches are also compromised. Strictly paralleling results on embryos, interfering with Hmga2 in a breast cancer cell model for EMT leads to molecular effects largely consistent with those observed on NCCs. These data indicate that Hmga2 is recruited in key molecular events that are shared by both NCCs and tumour cells.
Collapse
Affiliation(s)
- Simone Macrì
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy
| | - Luca Simula
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy
| | - Ilenia Pellarin
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Silvia Pegoraro
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Marco Onorati
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy
| | - Riccardo Sgarra
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Guidalberto Manfioletti
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Robert Vignali
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy.
| |
Collapse
|
9
|
Zhao Y, Shi J, Winey M, Klymkowsky MW. Identifying domains of EFHC1 involved in ciliary localization, ciliogenesis, and the regulation of Wnt signaling. Dev Biol 2016; 411:257-265. [PMID: 26783883 DOI: 10.1016/j.ydbio.2016.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/16/2015] [Accepted: 01/07/2016] [Indexed: 12/24/2022]
Abstract
EFHC1 encodes a ciliary protein that has been linked to Juvenile Myoclonic Epilepsy. In ectodermal explants, derived from Xenopus laevis embryos, the morpholino-mediated down-regulation of EFHC1b inhibited multiciliated cell formation. In those ciliated cells that did form, axoneme but not basal body formation was inhibited. EFHC1b morphant embryos displayed defects in central nervous system (CNS) and neural crest patterning that were rescued by a EFHC1b-GFP chimera. EFHC1b-GFP localized to ciliary axonemes in epidermal, gastrocoele roof plate, and neural tube cells. In X. laevis there is a link between Wnt signaling and multiciliated cell formation. While down-regulation of EFHC1b led to a ~2-fold increase in the activity of the β-catenin/Wnt-responsive TOPFLASH reporter, EFHC1b-GFP did not inhibit β-catenin activation of TOPFLASH. Wnt8a RNA levels were increased in EFHC1b morphant ectodermal explants and intact embryos, analyzed prior to the on-set of ciliogenesis. Rescue of the EFHC1b MO's ciliary axonemal phenotypes required the entire protein; in contrast, the EFHC1b morpholino's Wnt8a, CNS, and neural crest phenotypes were rescued by a truncated form of EFHC1b. The EFHC1b morpholino's Wnt8a phenotype was also rescued by the injection of RNAs encoding secreted Wnt inhibitors, suggesting that these phenotypes are due to effects on Wnt signaling, rather than the loss of cilia, an observation of potential relevance to understanding EFHC1's role in human neural development.
Collapse
Affiliation(s)
- Ying Zhao
- Molecular, Cellular & Developmental Biology, University of Colorado Boulder Boulder, Colorado 80309-0347, USA
| | - Jianli Shi
- Molecular, Cellular & Developmental Biology, University of Colorado Boulder Boulder, Colorado 80309-0347, USA
| | - Mark Winey
- Molecular, Cellular & Developmental Biology, University of Colorado Boulder Boulder, Colorado 80309-0347, USA
| | - Michael W Klymkowsky
- Molecular, Cellular & Developmental Biology, University of Colorado Boulder Boulder, Colorado 80309-0347, USA.
| |
Collapse
|
10
|
Lin C, Xin S, Qin X, Li H, Lin L, You Y. Zoledronic acid suppresses metastasis of esophageal squamous cell carcinoma cells through upregulating the tight junction protein occludin. Cytotechnology 2015. [PMID: 26204820 DOI: 10.1007/s10616-015-9884-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
We have previously demonstrated the radio-sensitizing effect of zoledronic acid (ZOL), a third generation bisphosphonate, on human esophageal squamous cell carcinoma (ESCC) cells. Here we show that ZOL suppresses metastatic progression of ESCC cells mainly through up-regulating the tight junction protein occludin. Exposure to ZOL at lower concentrations dramatically reduced migration and invasion of ESCC cells. In addition, ZOL treatment decreased the expression of mesenchymal markers, vimentin and N-cadherin, while increased the expression of the tight junction protein occludin. Moreover, ectopic expression of Slug, a well-known transcriptional repressor of occludin, partially but significantly abrogated the effect of ZOL on occludin expression and subsequently rescued the malignant metastatic phenotype, suggesting that Slug is one of the mediators underlying the anti-metastatic effect of ZOL. The present study is the first to report the significance of ZOL on ESCC metastasis. These data are promising for the future application of this drug regimen in patients with ESCC.
Collapse
Affiliation(s)
- Canfeng Lin
- Department of Radiation Oncology, Shantou Central Hospital, Shantou, 515041, China
| | - Shubo Xin
- Department of Clinical Pharmacy, Shantou Central Hospital, Shantou, 515041, China
| | - Xin Qin
- Medical College, Hubei University of Arts and Science, Xiangyang, 441053, China
| | - Haijun Li
- Department of Radiation Oncology, The Second People's Hospital of Neijiang City, Neijiang, 641000, China
| | - Lianxing Lin
- Department of Radiation Oncology, Shantou Central Hospital, Shantou, 515041, China
| | - Yanjie You
- Department of Pharmacy, Luohe Medical College, 148 Daxue-Road, Luohe, 462002, China.
| |
Collapse
|
11
|
Choi MJ, Cho KH, Lee S, Bae YJ, Jeong KJ, Rha SY, Choi EJ, Park JH, Kim JM, Lee JS, Mills GB, Lee HY. hTERT mediates norepinephrine-induced Slug expression and ovarian cancer aggressiveness. Oncogene 2015; 34:3402-12. [PMID: 25151968 DOI: 10.1038/onc.2014.270] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 07/07/2014] [Accepted: 07/15/2014] [Indexed: 12/13/2022]
Abstract
Stress hormones have been implicated in both tumor initiation and progression. Human telomerase reverse transcriptase (hTERT) is overexpressed in cancer cells and associated with malignant tumor progression and poor outcome. We thus sought to determine whether the stress hormone norepinephrine (NE) could induce hTERT expression and subsequently ovarian cancer progression. Unexpectedly, NE induced hTERT transcript and protein expression, and subsequently ovarian cancer cell invasion. Pharmacologic inhibition of β2-adrenergic receptor 2 and protein kinase A, as well as silencing of hypoxia-inducible factor-1α and c-Myc expression, profoundly attenuated NE-induced hTERT expression. Strikingly, stimulation of the cells with NE or ectopic expression of hTERT induced expression of Slug, ovarian cancer cell epithelial-mesenchymal transition (EMT) and invasion. Silencing of hTERT expression abrogated NE-induced ovarian cancer cell invasion, EMT and Slug expression. In addition, silencing of Slug expression significantly inhibited NE- and hTERT-induced ovarian cancer cell EMT and invasion. Moreover, continuous exposure to NE was sufficient to enhance in vivo hTERT expression and metastasis of ovarian cancer cells to the lung. Finally, we provide evidence that hTERT links Src to Slug expression in NE-induced ovarian cancer EMT and metastasis. We thus demonstrate a novel role of hTERT in stress hormone-induced ovarian cancer aggressiveness through inducing Slug, providing novel biomarkers and potential therapeutic targets for ovarian cancer.
Collapse
Affiliation(s)
- M J Choi
- Department of Pharmacology, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, Korea
| | - K H Cho
- Department of Pharmacology, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, Korea
| | - S Lee
- Department of Biochemistry, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, Korea
| | - Y J Bae
- Department of Pharmacology, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, Korea
| | - K J Jeong
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S Y Rha
- Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - E J Choi
- Department of Optometry, College of Medical Science, Konyang University, Daejeon, Korea
| | - J H Park
- Department of Biochemistry, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, Korea
| | - J M Kim
- Cancer Research Institute, Regional Cancer Center and Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - J-S Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - H Y Lee
- 1] Department of Pharmacology, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, Korea [2] Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
12
|
Tien CL, Jones A, Wang H, Gerigk M, Nozell S, Chang C. Snail2/Slug cooperates with Polycomb repressive complex 2 (PRC2) to regulate neural crest development. Development 2015; 142:722-31. [PMID: 25617436 PMCID: PMC4325378 DOI: 10.1242/dev.111997] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 12/11/2014] [Indexed: 12/25/2022]
Abstract
Neural crest cells arise from the border of the neural plate and epidermal ectoderm, migrate extensively and differentiate into diverse cell types during vertebrate embryogenesis. Although much has been learnt about growth factor signals and gene regulatory networks that regulate neural crest development, limited information is available on how epigenetic mechanisms control this process. In this study, we show that Polycomb repressive complex 2 (PRC2) cooperates with the transcription factor Snail2/Slug to modulate neural crest development in Xenopus. The PRC2 core components Eed, Ezh2 and Suz12 are expressed in the neural crest cells and are required for neural crest marker expression. Knockdown of Ezh2, the catalytic subunit of PRC2 for histone H3K27 methylation, results in defects in neural crest specification, migration and craniofacial cartilage formation. EZH2 interacts directly with Snail2, and Snail2 fails to expand the neural crest domains in the absence of Ezh2. Chromatin immunoprecipitation analysis shows that Snail2 regulates EZH2 occupancy and histone H3K27 trimethylation levels at the promoter region of the Snail2 target E-cadherin. Our results indicate that Snail2 cooperates with EZH2 and PRC2 to control expression of the genes important for neural crest specification and migration during neural crest development.
Collapse
Affiliation(s)
- Chih-Liang Tien
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue S., Birmingham, AL 35294, USA
| | - Amanda Jones
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1720 2nd Avenue S., Birmingham, AL 35294, USA
| | - Hengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1720 2nd Avenue S., Birmingham, AL 35294, USA
| | - Magda Gerigk
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue S., Birmingham, AL 35294, USA
| | - Susan Nozell
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue S., Birmingham, AL 35294, USA
| | - Chenbei Chang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue S., Birmingham, AL 35294, USA
| |
Collapse
|
13
|
Lindsey S, Langhans SA. Crosstalk of Oncogenic Signaling Pathways during Epithelial-Mesenchymal Transition. Front Oncol 2014; 4:358. [PMID: 25566498 PMCID: PMC4263086 DOI: 10.3389/fonc.2014.00358] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 11/27/2014] [Indexed: 12/11/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) and cell transformation have been well-documented in multiple cancer cell models and are believed to be one of the earliest events in tumor progression. Genetic and epigenetic modifications shift cells toward either end of the EMT spectrum, and can be influenced by the microenvironment surrounding a tumor. EMT and mesenchymal–epithelial transition are critical to normal function and development and an intricate network of transcription factors and transcriptional regulators tightly regulates these processes. As evidenced in normal and transformed cell lines, many signaling pathways trigger EMT during development and differentiation. The signaling pathways include those triggered by different members of the transforming growth factor superfamily, epidermal growth factor, fibroblast growth factor, hepatocyte growth factor, hypoxia-inducible factor, Wnt, Notch, and many others. Functional redundancies allow cells to undergo EMT even if these key transcriptional regulators are lacking, but these same redundancies also make these pathways particularly susceptible to gain-of-function mutations or constitutive signal activation; the “forced” transition toward either a mesenchymal or epithelial phenotype.
Collapse
Affiliation(s)
- Stephan Lindsey
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | - Sigrid A Langhans
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| |
Collapse
|
14
|
Bisson N, Wedlich D, Moss T. The p21-activated kinase Pak1 regulates induction and migration of the neural crest in Xenopus. Cell Cycle 2014; 11:1316-24. [DOI: 10.4161/cc.19685] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
15
|
Chibby functions in Xenopus ciliary assembly, embryonic development, and the regulation of gene expression. Dev Biol 2014; 395:287-98. [PMID: 25220153 DOI: 10.1016/j.ydbio.2014.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 12/21/2022]
Abstract
Wnt signaling and ciliogenesis are core features of embryonic development in a range of metazoans. Chibby (Cby), a basal-body associated protein, regulates β-catenin-mediated Wnt signaling in the mouse but not Drosophila. Here we present an analysis of Cby's embryonic expression and morphant phenotypes in Xenopus laevis. Cby RNA is supplied maternally, negatively regulated by Snail2 but not Twist1, preferentially expressed in the neuroectoderm, and regulates β-catenin-mediated gene expression. Reducing Cby levels reduced the density of multiciliated cells, the number of basal bodies per multiciliated cell, and the numbers of neural tube primary cilia; it also led to abnormal development of the neural crest, central nervous system, and pronephros, all defects that were rescued by a Cby-GFP chimera. Reduction of Cby led to an increase in Wnt8a and decreases in Gli2, Gli3, and Shh RNA levels. Many, but not all, morphant phenotypes were significantly reversed by the Wnt inhibitor SFRP2. These observations extend our understanding of Cby's role in mediating the network of interactions between ciliogenesis, signaling systems and tissue patterning.
Collapse
|
16
|
Sakamaki K, Takagi C, Kitayama A, Kurata T, Yamamoto TS, Chiba K, Kominami K, Jung SK, Okawa K, Nozaki M, Kubota HY, Ueno N. Multiple functions of FADD in apoptosis, NF-κB-related signaling, and heart development in Xenopus embryos. Genes Cells 2012; 17:875-96. [PMID: 23025414 DOI: 10.1111/gtc.12004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 08/19/2012] [Indexed: 12/26/2022]
Abstract
FADD is an adaptor protein that transmits apoptotic signals from death receptors. Additionally, FADD has been shown to play a role in various functions including cell proliferation. However, the physiological role of FADD during embryonic development remains to be delineated. Here, we show the novel roles FADD plays in development and the molecular mechanisms of these roles in Xenopus embryos. By whole-mount in situ hybridization and RT-PCR analysis, we observed that fadd is constantly expressed in early embryos. The upregulation or downregulation of FADD proteins by embryonic manipulation resulted in induction of apoptosis or size changes in the heart during development. Expression of a truncated form of FADD, FADDdd, which lacks pro-apoptotic activity, caused growth retardation of embryos associated with dramatic expressional fluctuations of genes that are regulated by NF-κB. Moreover, we isolated a homolog of mammalian cullin-4 (Cul4), a component of the ubiquitin E3 ligase family, as a FADDdd-interacting molecule in Xenopus embryos. Thus, our study shows that FADD has multiple functions in embryos; it plays a part in the regulation of NF-κB activation and heart formation, in addition to apoptosis. Furthermore, our findings provide new insights into how Cul4-based ligase is related to FADD signaling in embryogenesis.
Collapse
Affiliation(s)
- Kazuhiro Sakamaki
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Huin-Schohn C, Guéguinou N, Schenten V, Bascove M, Koch GG, Baatout S, Tschirhart E, Frippiat JP. Gravity changes during animal development affect IgM heavy-chain transcription and probably lymphopoiesis. FASEB J 2012; 27:333-41. [PMID: 22993194 DOI: 10.1096/fj.12-217547] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our previous research demonstrated that spaceflight conditions affect antibody production in response to an antigenic stimulation in adult amphibians. Here, we investigated whether antibody synthesis is affected when animal development occurs onboard a space station. To answer this question, embryos of the Iberian ribbed newt, Pleurodeles waltl, were sent to the International Space Station (ISS) before the initiation of immunoglobulin heavy-chain expression. Thus, antibody synthesis began in space. On landing, we determined the effects of spaceflight on P. waltl development and IgM heavy-chain transcription. Results were compared with those obtained using embryos that developed on Earth. We find that IgM heavy-chain transcription is doubled at landing and that spaceflight does not affect P. waltl development and does not induce inflammation. We also recreated the environmental modifications encountered by the embryos during their development onboard the ISS. This strategy allowed us to demonstrate that gravity change is the factor responsible for antibody heavy-chain transcription modifications that are associated with NF-κB mRNA level variations. Taken together, and given that the larvae were not immunized, these data suggest a modification of lymphopoiesis when gravity changes occur during ontogeny.
Collapse
Affiliation(s)
- Cécile Huin-Schohn
- Department of Development and Immunogenetics, Lorraine University, Vandoeuvre-lès-Nancy, France
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Kerosuo L, Bronner-Fraser M. What is bad in cancer is good in the embryo: importance of EMT in neural crest development. Semin Cell Dev Biol 2012; 23:320-32. [PMID: 22430756 PMCID: PMC3345076 DOI: 10.1016/j.semcdb.2012.03.010] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 02/14/2012] [Accepted: 03/01/2012] [Indexed: 11/18/2022]
Abstract
Although the epithelial to mesenchymal transition (EMT) is famous for its role in cancer metastasis, it also is a normal developmental event in which epithelial cells are converted into migratory mesenchymal cells. A prime example of EMT during development occurs when neural crest (NC) cells emigrate from the neural tube thus providing an excellent model to study the principles of EMT in a nonmalignant environment. NC cells start life as neuroepithelial cells intermixed with precursors of the central nervous system. After EMT, they delaminate and begin migrating, often to distant sites in the embryo. While proliferating and maintaining multipotency and cell survival the transitioning neural crest cells lose apicobasal polarity and the basement membrane is broken down. This review discusses how these events are coordinated and regulated, by series of events involving signaling factors, gene regulatory interactions, as well as epigenetic and post-transcriptional modifications. Even though the series of events involved in NC EMT are well known, the sequence in which these steps take place remains a subject of debate, raising the intriguing possibility that, rather than being a single event, neural crest EMT may involve multiple parallel mechanisms.
Collapse
Affiliation(s)
- Laura Kerosuo
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, United States
| | | |
Collapse
|
19
|
Shi J, Zhang H, Dowell RD, Klymkowsky MW. sizzled function and secreted factor network dynamics. Biol Open 2012; 1:286-94. [PMID: 23213419 PMCID: PMC3507283 DOI: 10.1242/bio.2012019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Studies on the role of the E-box binding transcription factor Snail2 (Slug) in the induction of neural crest by mesoderm (Shi et al., 2011) revealed an unexpected increase in the level of sizzled RNA in the dorsolateral mesodermal zone (DMLZ) of morphant Xenopus embryos. sizzled encodes a secreted protein with both Wnt and BMP inhibitor activities. Morpholino-mediated down-regulation of sizzled expression in one cell of two cell embryos or the C2/C3 blastomeres of 32-cell embryos, which give rise to the DLMZ, revealed decreased expression of the mesodermal marker brachyury and subsequent defects in neural crest induction, pronephros formation, and muscle patterning. Loss of sizzled expression led to decreases in RNAs encoding the secreted Wnt inhibitor SFRP2 and the secreted BMP inhibitor Noggin; the sizzled morphant phenotype could be rescued by co-injection of RNAs encoding Noggin and either SFRP2 or Dickkopf (a mechanistically distinct Wnt inhibitor). Together, these observations reveal that sizzled, in addition to its established role in dorsal-ventral patterning, is also part of a dynamic BMP and Wnt signaling network involved in both mesodermal patterning and neural crest induction.
Collapse
Affiliation(s)
- Jianli Shi
- Molecular, Cellular and Developmental Biology, University of Colorado , Boulder, CO 80309-0347 , USA
| | | | | | | |
Collapse
|
20
|
Wei S, Xu G, Bridges LC, Williams P, Nakayama T, Shah A, Grainger RM, White JM, DeSimone DW. Roles of ADAM13-regulated Wnt activity in early Xenopus eye development. Dev Biol 2011; 363:147-54. [PMID: 22227340 DOI: 10.1016/j.ydbio.2011.12.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 12/13/2011] [Accepted: 12/19/2011] [Indexed: 10/14/2022]
Abstract
Pericellular proteolysis by ADAM family metalloproteinases has been widely implicated in cell signaling and development. We recently found that Xenopus ADAM13, an ADAM metalloproteinase, is required for activation of canonical Wnt signaling during cranial neural crest (CNC) induction by regulating a novel crosstalk between Wnt and ephrin B (EfnB) signaling pathways (Wei et al., 2010b). In the present study we show that the metalloproteinase activity of ADAM13 also plays important roles in eye development in Xenopus tropicalis. Knockdown of ADAM13 results in reduced expression of eye field markers pax6 and rx1, as well as that of the pan-neural marker sox2. Activation of canonical Wnt signaling or inhibition of forward EfnB signaling rescues the eye defects caused by loss of ADAM13, suggesting that ADAM13 functions through regulation of the EfnB-Wnt pathway interaction. Downstream of Wnt, the head inducer Cerberus was identified as an effector that mediates ADAM13 function in early eye field formation. Furthermore, ectopic expression of the Wnt target gene snail2 restores cerberus expression and rescues the eye defects caused by ADAM13 knockdown. Together these data suggest an important role of ADAM13-regulated Wnt activity in eye development in Xenopus.
Collapse
Affiliation(s)
- Shuo Wei
- Department of Cell Biology and the Morphogenesis and Regenerative Medicine Institute, University of Virginia Health System, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Klymkowsky M. Mitochondrial activity, embryogenesis, and the dialogue between the big and little brains of the cell. Mitochondrion 2011; 11:814-9. [DOI: 10.1016/j.mito.2010.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 11/02/2010] [Accepted: 11/18/2010] [Indexed: 12/31/2022]
|
22
|
Shi J, Severson C, Yang J, Wedlich D, Klymkowsky MW. Snail2 controls mesodermal BMP/Wnt induction of neural crest. Development 2011; 138:3135-45. [PMID: 21715424 DOI: 10.1242/dev.064394] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The neural crest is an induced tissue that is unique to vertebrates. In the clawed frog Xenopus laevis, neural crest induction depends on signals secreted from the prospective dorsolateral mesodermal zone during gastrulation. The transcription factors Snail2 (Slug), Snail1 and Twist1 are expressed in this region. It is known that Snail2 and Twist1 are required for both mesoderm formation and neural crest induction. Using targeted blastomere injection, morpholino-based loss of function and explant studies, we show that: (1) Snail1 is also required for mesoderm and neural crest formation; (2) loss of snail1, snail2 or twist1 function in the C2/C3 lineage of 32-cell embryos blocks mesoderm formation, but neural crest is lost only in the case of snail2 loss of function; (3) snail2 mutant loss of neural crest involves mesoderm-derived secreted factors and can be rescued synergistically by bmp4 and wnt8 RNAs; and (4) loss of snail2 activity leads to changes in the RNA levels of a number of BMP and Wnt agonists and antagonists. Taken together, these results identify Snail2 as a key regulator of the signals involved in mesodermal induction of neural crest.
Collapse
Affiliation(s)
- Jianli Shi
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309-0347, USA
| | | | | | | | | |
Collapse
|
23
|
Xrel3/XrelA attenuates β-catenin-mediated transcription during mesoderm formation in Xenopus embryos. Biochem J 2011; 435:247-57. [PMID: 21214516 DOI: 10.1042/bj20101801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In Xenopus laevis embryonic development, activation of the Wnt/β-catenin pathway promotes mesoderm cell fate determination via Xnr (Xenopus nodal-related) expression. We have demonstrated previously that Rel/NF-κB (nuclear factor κB) proteins expressed in presumptive ectoderm limit the activity of Xnrs to the marginal zone of embryos during mesoderm induction, which assists to distinguish mesoderm from ectoderm. The mechanism of this regulation, however, is unknown. In the present study, we investigated whether Rel/NF-κB proteins are able to modulate mesoderm formation by mediating Wnt/β-catenin signalling. We determined that ectopic expression of XrelA or Xrel3 in the dorsal marginal zone perturbed dorsal mesoderm formation by down-regulating multiple Wnt/β-catenin target genes including Xnr3, Xnr5 and Xnr6. Ventral co-expression of XrelA or Xrel3 with either wild-type β-catenin or constitutively active β-cateninS37A abrogated β-catenin-induced axis duplication and attenuated β-catenin-stimulated reporter transcription. Lastly, we provide evidence that Xrel3, but not XrelA, can interact with β-catenin without affecting the association of β-catenin with other transcriptional co-activators in vitro. Both Xrel3 and XrelA, however, prevented the accumulation, in nuclei, of exogenously expressed and endogenous β-catenin in vivo. These results suggest that Rel proteins are able to bind β-catenin and attenuate β-catenin-mediated transcription by nuclear exclusion.
Collapse
|
24
|
Tsuchiya Y, Yamashita S. Anti-apoptotic activity and proteasome-mediated degradation of Xenopus Mcl-1 protein in egg extracts. J Biol Chem 2011; 286:15806-14. [PMID: 21454490 DOI: 10.1074/jbc.m110.175927] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Xenopus egg extracts execute spontaneous apoptosis without the requirement of transcription and translation, and this intrinsic mechanism is supposed to be involved in the physiological elimination of aged eggs. Although apoptosis in this system is carried out by maternally stockpiled materials, the endogenous apoptosis regulators present in egg extracts are still poorly characterized. Here we examined the mRNA expression profiles and apoptosis-regulating functions of 13 Xenopus Bcl-2 family proteins in egg extracts. Among these, we found that endogenous Xenopus Mcl-1 (xMcl-1) physiologically inhibited apoptosis by counteracting the pro-apoptotic activity of endogenous Xenopus Bid in egg extracts. Exogenously added recombinant xMcl-1 was rapidly degraded by proteasome in egg extracts, and we identified the destabilizing region in the N terminus of xMcl-1. Our results suggest that the proteolytic decay of xMcl-1 may change the functional balance between pro- and anti-apoptotic activities of Bcl-2 family proteins, thereby regulating the timing of cytochrome c release in egg extracts.
Collapse
Affiliation(s)
- Yuichi Tsuchiya
- Department of Biochemistry, Toho University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
25
|
Franco DL, Mainez J, Vega S, Sancho P, Murillo MM, de Frutos CA, Del Castillo G, López-Blau C, Fabregat I, Nieto MA. Snail1 suppresses TGF-beta-induced apoptosis and is sufficient to trigger EMT in hepatocytes. J Cell Sci 2011; 123:3467-77. [PMID: 20930141 DOI: 10.1242/jcs.068692] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although TGF-β suppresses early stages of tumour development, it later contributes to tumour progression when cells become resistant to its suppressive effects. In addition to circumventing TGF-β-induced growth arrest and apoptosis, malignant tumour cells become capable of undergoing epithelial-to-mesenchymal transition (EMT), favouring invasion and metastasis. Therefore, defining the mechanisms that allow cancer cells to escape from the suppressive effects of TGF-β is fundamental to understand tumour progression and to design specific therapies. Here, we have examined the role of Snail1 as a suppressor of TGF-β-induced apoptosis in murine non-transformed hepatocytes, rat and human hepatocarcinoma cell lines and transgenic mice. We show that Snail1 confers resistance to TGF-β-induced cell death and that it is sufficient to induce EMT in adult hepatocytes, cells otherwise refractory to this transition upon exposure to TGF-β. Furthermore, we show that Snail1 silencing prevents EMT and restores the cell death response induced by TGF-β. As Snail1 is a known target of TGF-β signalling, our data indicate that Snail1 might transduce the tumour-promoting effects of TGF-β, namely the EMT concomitant with the resistance to cell death.
Collapse
Affiliation(s)
- D Lorena Franco
- Instituto de Neurociencias (CSIC-UMH), 03550 San Juan de Alicante, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Endo T, Kusakabe M, Sunadome K, Yamamoto T, Nishida E. The kinase SGK1 in the endoderm and mesoderm promotes ectodermal survival by down-regulating components of the death-inducing signaling complex. Sci Signal 2011; 4:ra2. [PMID: 21245468 DOI: 10.1126/scisignal.2001211] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
A balance between cell survival and apoptosis is essential for animal development. Although proper development involves multiple interactions between germ layers, little is known about the intercellular and intertissue signaling pathways that promote cell survival in neighboring or distant germ layers. We found that serum- and glucocorticoid-inducible kinase 1 (SGK1) promoted ectodermal cell survival during early Xenopus embryogenesis through a non-cell-autonomous mechanism. Dorsal depletion of SGK1 in Xenopus embryos resulted in shortened axes and reduced head structures with defective eyes, and ventral depletion led to defective tail morphologies. Although the gene encoding SGK1 was mainly expressed in the endoderm and dorsal mesoderm, knockdown of SGK1 caused excessive apoptosis in the ectoderm. SGK1-depleted ectodermal explants showed little or no apoptosis, suggesting non-cell-autonomous effects of SGK1 on ectodermal cells. Microarray analysis revealed that SGK1 knockdown increased the expression of genes encoding FADD (Fas-associated death domain protein) and caspase-10, components of the death-inducing signaling complex (DISC). Inhibition of DISC function suppressed excessive apoptosis in SGK1-knockdown embryos. SGK1 acted through the transcription factor nuclear factor κB (NF-κB) to stimulate production of bone morphogenetic protein 7 (BMP7), and overexpression of BMP7 in SGK1-knockdown embryos reduced the abundance of DISC components. We show that phosphoinositide 3-kinase (PI3K) functioned upstream of SGK1, thus revealing an endodermal and mesodermal pathway from PI3K to SGK1 to NF-κB that produces BMP7, which promotes ectodermal survival by decreasing DISC function.
Collapse
Affiliation(s)
- Tatsuya Endo
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | | | | | | | | |
Collapse
|
27
|
Schneider M, Schambony A, Wedlich D. Prohibitin1 acts as a neural crest specifier in Xenopus development by repressing the transcription factor E2F1. Development 2010; 137:4073-81. [PMID: 21062864 DOI: 10.1242/dev.053405] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Prohibitin 1 (phb1), which was initially described as an inhibitor of cell proliferation, is a highly conserved protein found in multiple cellular compartments. In the nucleus it interacts with the transcriptional regulators Rb and E2F1 and controls cell proliferation and apoptosis. Here we unravel an unexpected novel function for phb1 in Xenopus cranial neural crest (CNC) development. Xphb1 is maternally expressed; zygotically expressed neurula stage transcripts accumulate in the CNC and the neural tube. Knockdown of Xphb1 by antisense morpholino injection results in the loss of foxD3, snail2 and twist expression, whereas expression of c-myc, AP-2 and snail1 remains unaffected. Xphb2, its closest relative, cannot substitute for Xphb1, underlining the specificity of Xphb1 function. Epistatic analyses place Xphb1 downstream of c-myc and upstream of foxD3, snail2 and twist. To elucidate which subdomain in Xphb1 is required for neural crest gene regulation we generated deletion mutants and tested their rescue ability in Xphb1 morphants. The E2F1-binding domain was found to be necessary for Xphb1 function in neural crest development. Gain- and loss-of-function experiments reveal that Xphb1 represses E2F1 activity; suppression of E2F1 through Xphb1 is required for twist, snail2 and foxD3 expression in the CNC. With the Xphb1 dependency of a subset of CNC specifiers downstream of c-myc, we have identified a new branching point in the neural crest gene regulatory network.
Collapse
Affiliation(s)
- Martina Schneider
- KIT, Campus South, Zoologicak Institute, Cell and Developmental Biology, Kaiserstrasse 12, Karlsruhe, Germany
| | | | | |
Collapse
|
28
|
Abstract
The epithelial-mesenchymal transition (EMT) describes a rapid and often reversible modulation of phenotype by epithelial cells. EMT was originally defined in the context of developmental stages, including heart morphogenesis, mesoderm and neural crest formation. Epithelial cells loosen cell-cell adhesion structures throughout EMT. They modulate their polarity, cytoskeleton organization and typically express vimentin filaments and downregulate cytokeratins. They become isolated, mobile and resistant to anoikis. The EMT at least superficially resembles the evolution from normal to transformed cell phenotype during carcinoma progression. The relevance of the concept of EMT in this context was indicated by in vitro models using transformed epithelial cells. Transduction pathways typical of embryogenic EMT in vivo were also found to be activated during cancer progression. More recently, it has been found that such pathways indicate an increased plasticity linked to cellular stemness and ability to generate tumors. However, in the absence of direct evidence, a number of oncologists and pathologists remain skeptical about applying the EMT concept to human tumor progression. Typically in the cancer field, EMT concept appears to be fully relevant in some situations, but the concept has to be adjusted in other situations to reflect tumor cell renewal and plasticity during carcinoma progression and metastasis.
Collapse
Affiliation(s)
- P Savagner
- IRCM U896 INSERM, CRLC Val d'Aurelle-Paul Lamarque, Montpellier, France.
| |
Collapse
|
29
|
Klymkowsky MW, Rossi CC, Artinger KB. Mechanisms driving neural crest induction and migration in the zebrafish and Xenopus laevis. Cell Adh Migr 2010; 4:595-608. [PMID: 20962584 PMCID: PMC3011258 DOI: 10.4161/cam.4.4.12962] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 07/09/2010] [Indexed: 01/09/2023] Open
Abstract
The neural crest is an evolutionary adaptation, with roots in the formation of mesoderm. Modification of neural crest behavior has been is critical for the evolutionary diversification of the vertebrates and defects in neural crest underlie a range of human birth defects. There has been a tremendous increase in our knowledge of the molecular, cellular, and inductive interactions that converge on defining the neural crest and determining its behavior. While there is a temptation to look for simple models to explain neural crest behavior, the reality is that the system is complex in its circuitry. In this review, our goal is to identify the broad features of neural crest origins (developmentally) and migration (cellularly) using data from the zebrafish (teleost) and Xenopus laevis (tetrapod amphibian) in order to illuminate where general mechanisms appear to be in play, and equally importantly, where disparities in experimental results suggest areas of profitable study.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Department of Molecular, Cellular and Developmental Biology; University of Colorado Boulder; Boulder, CO USA
| | - Christy Cortez Rossi
- Department of Craniofacial Biology; University of Colorado Denver; School of Dental Medicine; Aurora, CO USA
| | - Kristin Bruk Artinger
- Department of Craniofacial Biology; University of Colorado Denver; School of Dental Medicine; Aurora, CO USA
| |
Collapse
|
30
|
Cha HS, Bae EK, Ahn JK, Lee J, Ahn KS, Koh EM. Slug suppression induces apoptosis via Puma transactivation in rheumatoid arthritis fibroblast-like synoviocytes treated with hydrogen peroxide. Exp Mol Med 2010; 42:428-36. [PMID: 20418652 PMCID: PMC2892596 DOI: 10.3858/emm.2010.42.6.044] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2010] [Indexed: 11/04/2022] Open
Abstract
Inadequate apoptosis contributes to synovial hyperplasia in rheumatoid arthritis (RA). Recent study shows that low expression of Puma might be partially responsible for the decreased apoptosis of fibroblast-like synoviocytes (FLS). Slug, a highly conserved zinc finger transcriptional repressor, is known to antagonize apoptosis of hematopoietic progenitor cells by repressing Puma transactivation. In this study, we examined the expression and function of Slug in RA FLS. Slug mRNA expression was measured in the synovial tissue (ST) and FLS obtained from RA and osteoarthritis patients. Slug and Puma mRNA expression in FLS by apoptotic stimuli were measured by real-time PCR analysis. FLS were transfected with control siRNA or Slug siRNA. Apoptosis was quantified by trypan blue exclusion, DNA fragmentation and caspase-3 assay. RA ST expressed higher level of Slug mRNA compared with osteoarthritis ST. Slug was significantly induced by hydrogen peroxide (H2O2) but not by exogenous p53 in RA FLS. Puma induction by H2O2 stimulation was significantly higher in Slug siRNA-transfected FLS compared with control siRNA-transfected FLS. After H2O2 stimulation, viable cell number was significantly lower in Slug siRNA-transfected FLS compared with control siRNA-transfected FLS. Apoptosis enhancing effect of Slug siRNA was further confirmed by ELISA that detects cytoplasmic histone-associated DNA fragments and caspase-3 assay. These data demonstrate that Slug is overexpressed in RA ST and that suppression of Slug gene facilitates apoptosis of FLS by increasing Puma transactivation. Slug may therefore represent a potential therapeutic target in RA.
Collapse
Affiliation(s)
- Hoon-Suk Cha
- Department of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | | | | | | | | | | |
Collapse
|
31
|
Klymkowsky MW. A guide to the productive poking, prodding and injection of cells. Development 2009. [DOI: 10.1242/dev.040352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Michael W. Klymkowsky
- Molecular, Cellular and Developmental Biology and CU Teach, University of Colorado, Boulder, Boulder, CO 80309-0347, USA
| |
Collapse
|
32
|
Fardin P, Ognibene M, Vanni C, De Santanna A, Varesio L, Eva A. Induction of epithelial mesenchimal transition and vasculogenesis in the lenses of Dbl oncogene transgenic mice. PLoS One 2009; 4:e7058. [PMID: 19759912 PMCID: PMC2739440 DOI: 10.1371/journal.pone.0007058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 08/17/2009] [Indexed: 01/06/2023] Open
Abstract
Background The Dbl family of proteins represents a large group of proto-oncogenes involved in cell growth regulation. The numerous domains that are present in many Dbl family proteins suggest that they act to integrate multiple inputs in complicated signaling networks involving the Rho GTPases. Alterations of the normal function of these proteins lead to pathological processes such as developmental disorders and neoplastic transformation. We generated transgenic mice introducing the cDNA of Dbl oncogene linked to the metallothionein promoter into the germ line of FVB mice and found that onco-Dbl expression in mouse lenses affected proliferation, migration and differentiation of lens epithelial cells. Results We used high density oligonucleotide microarray to define the transcriptional profile induced by Dbl in the lenses of 2 days, 2 weeks, and 6 weeks old transgenic mice. We observed modulation of genes encoding proteins promoting epithelial-mesenchymal transition (EMT), such as down-regulation of epithelial cell markers and up-regulation of fibroblast markers. Genes encoding proteins involved in the positive regulation of apoptosis were markedly down regulated while anti-apoptotic genes were strongly up-regulated. Finally, several genes encoding proteins involved in the process of angiogenesis were up-regulated. These observations were validated by histological and immunohistochemical examination of the transgenic lenses where vascularization can be readily observed. Conclusion Onco-Dbl expression in mouse lens correlated with modulation of genes involved in the regulation of EMT, apoptosis and vasculogenesis leading to disruption of the lens architecture, epithelial cell proliferation, and aberrant angiogenesis. We conclude that onco-Dbl has a potentially important, previously unreported, capacity to dramatically alter epithelial cell migration, replication, polarization and differentiation and to induce vascularization of an epithelial tissue.
Collapse
Affiliation(s)
- Paolo Fardin
- Laboratorio di Biologia Molecolare, Istituto G. Gaslini, Genova, Italy
| | - Marzia Ognibene
- Laboratorio di Biologia Molecolare, Istituto G. Gaslini, Genova, Italy
| | - Cristina Vanni
- Laboratorio di Biologia Molecolare, Istituto G. Gaslini, Genova, Italy
| | - Amleto De Santanna
- Sezione di Istologia, Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Luigi Varesio
- Laboratorio di Biologia Molecolare, Istituto G. Gaslini, Genova, Italy
| | - Alessandra Eva
- Laboratorio di Biologia Molecolare, Istituto G. Gaslini, Genova, Italy
- * E-mail:
| |
Collapse
|
33
|
Zhang C, Klymkowsky MW. Unexpected functional redundancy between Twist and Slug (Snail2) and their feedback regulation of NF-kappaB via Nodal and Cerberus. Dev Biol 2009; 331:340-9. [PMID: 19389392 PMCID: PMC2747320 DOI: 10.1016/j.ydbio.2009.04.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Revised: 04/08/2009] [Accepted: 04/09/2009] [Indexed: 10/20/2022]
Abstract
A NF-kappaB-Twist-Snail network controls axis and mesoderm formation in Drosophila. Using translation-blocking morpholinos and hormone-regulated proteins, we demonstrate the presence of an analogous network in the early Xenopus embryo. Loss of twist (twist1) function leads to a reduction of mesoderm and neural crest markers, an increase in apoptosis, and a decrease in snail1 (snail) and snail2 (slug) mRNA levels. Injection of snail2 mRNA rescues twist's loss of function phenotypes and visa versa. In the early embryo NF-kappaB/RelA regulates twist, snail2, and snail1 mRNA levels; similarly Nodal/Smad2 regulate twist, snail2, snail1, and relA RNA levels. Both Twist and Snail2 negatively regulate levels of cerberus RNA, which encodes a Nodal, bone morphogenic protein (BMP), and Wnt inhibitor. Cerberus's anti-Nodal activity inhibits NF-kappaB activity and decreases relA RNA levels. These results reveal both conserved and unexpected regulatory interactions at the core of a vertebrate's mesodermal specification network.
Collapse
Affiliation(s)
| | - Michael W. Klymkowsky
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Boulder, CO 80309-0347, U.S.A
| |
Collapse
|
34
|
Craig EA, Parker P, Camenisch TD. Size-dependent regulation of Snail2 by hyaluronan: its role in cellular invasion. Glycobiology 2009; 19:890-8. [PMID: 19451547 DOI: 10.1093/glycob/cwp064] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hyaluronan (HA) induces changes in cellular behavior that are crucial during both embryonic development and cancer progression. However, the biological effects of varying sizes of HA and the signal transduction mechanisms that these polymers may activate remain unclear. In this study, we demonstrate that pulse stimulation of mouse embryonic fibroblasts with high-molecular-weight (HMW) HA, but not HA of lower molecular sizes, leads to increases in Snail2 protein which are dependent on NFkappaB activity. Involvement of CD44, the main HA receptor, in these responses was determined by use of a CD44 blocking antibody and CD44 siRNA. Both the blockade and silencing of CD44 significantly abrogate the increases in nuclear factor kappaB (NFkappaB) activity and Snail2 protein following HMW-HA stimulation. Furthermore, we show that HMW-HA induces cellular invasion and that inhibition of CD44, Snail2, or NFkappaB significantly decreases this response. These studies elucidate a novel HA/Snail2 functional connection through CD44 and NFkappaB that is important for the induction of cellular invasion and is dependent on HA size.
Collapse
Affiliation(s)
- Evisabel A Craig
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | | | | |
Collapse
|
35
|
Klymkowsky MW, Savagner P. Epithelial-mesenchymal transition: a cancer researcher's conceptual friend and foe. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1588-93. [PMID: 19342369 PMCID: PMC2671246 DOI: 10.2353/ajpath.2009.080545] [Citation(s) in RCA: 392] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/16/2009] [Indexed: 12/27/2022]
Abstract
Epithelial-mesenchymal transition (EMT) describes a series of rapid changes in cellular phenotype. During EMT, epithelial cells down-modulate cell-cell adhesion structures, alter their polarity, reorganize their cytoskeleton, and become isolated, motile, and resistant to anoikis. The term EMT is often applied to distinct biological events as if it were a single conserved process, but in fact EMT-related processes can vary in intensity from a transient loss of cell polarity to the total cellular reprogramming, as found by transcriptional analysis. Based on clinical observations, it is more appropriate in most cases to describe the emergence of an EMT-like phenotype during tumor progression. Although EMT implies complete trans-differentiation, EMT-like emphasizes the intermediary phenotype associated with tumor cell renewal and adaptation to specific microenvironments. Here, we categorize the various EMT-like phenotypes found in human carcinomas that, depending on the tumor type, may or not represent analogous stages in tumor progression. We based these categories on the global tumor phenotype. The tumor microenvironment, which is associated with stromal reactions, hypoxia, paucity of nutrients, impaired differentiation, and activation of various EMT-associated pathways, modulates overall tumor phenotype and leads to tumor heterogeneity.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Department of Molecular, Cellular, and Development Biology, University of Colorado, Boulder, Colorado, USA
| | | |
Collapse
|
36
|
Klymkowsky MW, Savagner P. Epithelial-mesenchymal transition: a cancer researcher's conceptual friend and foe. THE AMERICAN JOURNAL OF PATHOLOGY 2009. [PMID: 19342369 DOI: 10.2353/ajpath.2009.080545.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Epithelial-mesenchymal transition (EMT) describes a series of rapid changes in cellular phenotype. During EMT, epithelial cells down-modulate cell-cell adhesion structures, alter their polarity, reorganize their cytoskeleton, and become isolated, motile, and resistant to anoikis. The term EMT is often applied to distinct biological events as if it were a single conserved process, but in fact EMT-related processes can vary in intensity from a transient loss of cell polarity to the total cellular reprogramming, as found by transcriptional analysis. Based on clinical observations, it is more appropriate in most cases to describe the emergence of an EMT-like phenotype during tumor progression. Although EMT implies complete trans-differentiation, EMT-like emphasizes the intermediary phenotype associated with tumor cell renewal and adaptation to specific microenvironments. Here, we categorize the various EMT-like phenotypes found in human carcinomas that, depending on the tumor type, may or not represent analogous stages in tumor progression. We based these categories on the global tumor phenotype. The tumor microenvironment, which is associated with stromal reactions, hypoxia, paucity of nutrients, impaired differentiation, and activation of various EMT-associated pathways, modulates overall tumor phenotype and leads to tumor heterogeneity.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Department of Molecular, Cellular, and Development Biology, University of Colorado, Boulder, Colorado, USA
| | | |
Collapse
|
37
|
Du F, Yang R, Ma HL, Wang QY, Wei SL. Expression of transcriptional repressor Slug gene in mouse endometrium and its effect during embryo implantation. Appl Biochem Biotechnol 2009; 157:346-55. [PMID: 19172233 DOI: 10.1007/s12010-008-8521-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 12/29/2008] [Indexed: 11/27/2022]
Abstract
Slug, a member of the Snail family of zinc-finger transcription factors, is involved in regulating embryonic development and tumorigenesis. The aim of this study was to investigate the expression of Slug in mouse endometrium during early pregnancy and its possible role during embryo implantation. Fluorescence quantitative polymerase chain reaction and immunohistochemistry were applied to detect Slug mRNA and Slug protein expression in endometrium of nonpregnant and early pregnant mice, respectively. The expressions of Slug mRNA and its protein in pregnant group were higher than that in nonpregnant group and gradually increased from pregnancy day 1, reaching its maximum level on day 4 and then declining on days 5, 6, and 7. Immunohistochemistry showed that Slug protein was mainly present in luminal epithelium from pregnancy days 2 to 5 and in glandular epithelium from days 2 to 6 and enhanced significantly in stromal cells on days 4, 5, and 6. The number of embryos implanted was greatly decreased after Slug function in mouse endometrium was blocked by the intrauterine injection with anti-Slug polyclonal antibody on day 3 of pregnancy before implantation. These results suggested that up-regulation of Slug expression may play a key role in the embryo implantation in mice.
Collapse
Affiliation(s)
- Fang Du
- Department of the Reproductive Physiology, College of Public Health, Chongqing Medical University, Chongqing 400016, China
| | | | | | | | | |
Collapse
|
38
|
Gabellini C, Castellini L, Trisciuoglio D, Kracht M, Zupi G, Del Bufalo D. Involvement of nuclear factor-kappa B in bcl-xL-induced interleukin 8 expression in glioblastoma. J Neurochem 2008; 107:871-82. [PMID: 18786178 DOI: 10.1111/j.1471-4159.2008.05661.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We recently reported that bcl-xL regulates interleukin 8 (CXCL8) protein expression and promoter activity in glioblastoma cells. In this paper we demonstrate that CXCL8 induction by bcl-xL is mediated through a nuclear factor-kappa B (NF-kB)-dependent mechanism. Mutational studies on the CXCL8 promoter showed that NF-kB binding site was required for bcl-xL-induced promoter activity and an enhanced nuclear expression of NF-kB subunits p65 and p50 was observed after bcl-xL over-expression. Electrophoretic mobility shift assay showed an increased DNA-binding activity of NF-kB in bcl-xL over-expressing cells and the use of specific antibodies confirmed the involvement of p65 and p50 in NF-kB activity on CXCL8 promoter sequence. NF-kB activity regulation by bcl-xL involved IkBalpha and IKK complex signaling pathway. In fact, bcl-xL over-expression induced a decrease of cytoplasmic expression of the IkBalpha protein, paralleled by an increase in the phosphorylation of the same IkBalpha and IKKalpha/beta. Moreover, the down-regulation of the ectopic or endogenous bcl-xL expression through RNA interference confirmed the ability of bcl-xL to modulate NF-kB pathway, and the transient expression of a degradation-resistant form of the cytoplasmic NF-kB inhibitor IkBalpha in bcl-xL transfectants confirmed the involvement of that inhibitor in bcl-xL-induced CXCL8 expression and promoter activity. In conclusion, our results demonstrate the role of NF-kB as the mediator of bcl-xL-induced CXCL8 up-regulation in glioblastoma cells.
Collapse
Affiliation(s)
- Chiara Gabellini
- Experimental Chemotherapy Laboratory, Regina Elena Cancer Institute, Rome, Italy
| | | | | | | | | | | |
Collapse
|
39
|
Nunes da Fonseca R, von Levetzow C, Kalscheuer P, Basal A, van der Zee M, Roth S. Self-regulatory circuits in dorsoventral axis formation of the short-germ beetle Tribolium castaneum. Dev Cell 2008; 14:605-15. [PMID: 18410735 DOI: 10.1016/j.devcel.2008.02.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Revised: 12/14/2007] [Accepted: 02/13/2008] [Indexed: 01/12/2023]
Abstract
The rel/NF-kappaB transcription factor Dorsal controls dorsoventral (DV) axis formation in Drosophila. A stable nuclear gradient of Dorsal directly regulates approximately 50 target genes. In Tribolium castaneum (Tc), a beetle with an ancestral type of embryogenesis, the Dorsal nuclear gradient is not stable, but rapidly shrinks and disappears. We find that negative feedback accounts for this dynamic behavior: Tc-Dorsal and one of its target genes activate transcription of the IkB homolog Tc-cactus, terminating Dorsal function. Despite its transient role, Tc-Dorsal is strictly required to initiate DV polarity, as in Drosophila. However, unlike in Drosophila, embryos lacking Tc-Dorsal display a periodic pattern of DV cell fates along the AP axis, indicating that a self-organizing ectodermal patterning system operates independently of mesoderm or maternal DV polarity cues. Our results also elucidate how extraembryonic tissues are organized in short-germ embryos, and how patterning information is transmitted from the early embryo to the growth zone.
Collapse
Affiliation(s)
- Rodrigo Nunes da Fonseca
- Institute of Developmental Biology, University of Cologne, Gyrhofstrasse 17, 50931 Cologne, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Langer EM, Feng Y, Zhaoyuan H, Rauscher FJ, Kroll KL, Longmore GD. Ajuba LIM proteins are snail/slug corepressors required for neural crest development in Xenopus. Dev Cell 2008; 14:424-36. [PMID: 18331720 DOI: 10.1016/j.devcel.2008.01.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Revised: 11/20/2007] [Accepted: 01/09/2008] [Indexed: 01/05/2023]
Abstract
Snail family transcriptional repressors regulate epithelial mesenchymal transitions during physiological and pathological processes. A conserved SNAG repression domain present in all vertebrate Snail proteins is necessary for repressor complex assembly. Here, we identify the Ajuba family of LIM proteins as functional corepressors of the Snail family via an interaction with the SNAG domain. Ajuba LIM proteins interact with Snail in the nucleus on endogenous E-cadherin promoters and contribute to Snail-dependent repression of E-cadherin. Using Xenopus neural crest as a model of in vivo Snail- or Slug-induced EMT, we demonstrate that Ajuba LIM proteins contribute to neural crest development as Snail/Slug corepressors and are required for in vivo Snail/Slug function. Because Ajuba LIM proteins are also components of adherens junctions and contribute to their assembly or stability, their functional interaction with Snail proteins in the nucleus suggests that Ajuba LIM proteins are important regulators of epithelia dynamics communicating surface events with nuclear responses.
Collapse
Affiliation(s)
- Ellen M Langer
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
41
|
Acloque H, Thiery JP, Nieto MA. The physiology and pathology of the EMT. Meeting on the epithelial-mesenchymal transition. EMBO Rep 2008; 9:322-6. [PMID: 18323854 PMCID: PMC2288772 DOI: 10.1038/embor.2008.30] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Accepted: 02/07/2008] [Indexed: 12/21/2022] Open
Affiliation(s)
- Hervé Acloque
- Instituto de Neurociencias de Alicante, CSIC-UMH, Apartado 18, San Juan de Alicante, 03550 Spain
| | - Jean Paul Thiery
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, 138673 Singapore
| | - M. Angela Nieto
- Instituto de Neurociencias de Alicante, CSIC-UMH, Apartado 18, San Juan de Alicante, 03550 Spain
| |
Collapse
|
42
|
Kennedy MW, Green KA, Ford RL, Andrews PG, Paterno GD, Gillespie LL, Kao KR. Regulation of the response to Nodal-mediated mesoderm induction by Xrel3. Dev Biol 2007; 311:383-95. [PMID: 17920056 DOI: 10.1016/j.ydbio.2007.08.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 08/20/2007] [Accepted: 08/22/2007] [Indexed: 11/15/2022]
Abstract
The Xenopus egg has a yolk-laden vegetal hemisphere juxtaposed to a darkly pigmented animal hemisphere. Mesoderm is derived from the marginal zone, located at the interface between the two hemispheres. The vegetal-most cells become endoderm and release TGF-beta-related factors, including the Xenopus Nodal related (Xnr) proteins, which diffuse to induce the marginal zone to form mesoderm. The remaining animal cells become ectoderm, but our understanding of the mechanisms that limit the response to induction is incomplete. In this study, we provide evidence to suggest that Xrel3, a member of the Rel/NF-kappaB family, plays a role in defining the boundary separating induced from uninduced cells by regulating Xnr-responsive gene transcription. Ectopic Xrel3 expressed in prospective mesoderm caused repression of mesoderm-specific genes resulting in loss-of-function phenotypes that were rescued by co-expression of Xnr2. Depletion of Xrel3 from embryos with antisense morpholinos increased Xnr-dependent transcription, broadened expression of the pan-mesoderm marker Xbra and sensitized animal cells to mesoderm induction by Xnr2. We propose that an additional component to the mechanism that differentiates the ectoderm from the mesoderm involves regulation of nodal-dependent gene transcription by Xrel3.
Collapse
Affiliation(s)
- Mark W Kennedy
- Terry Fox Cancer Research Labs, Memorial University of Newfoundland, St. John's, NL, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Taylor KM, LaBonne C. Modulating the activity of neural crest regulatory factors. Curr Opin Genet Dev 2007; 17:326-31. [PMID: 17651964 DOI: 10.1016/j.gde.2007.05.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Revised: 05/24/2007] [Accepted: 05/31/2007] [Indexed: 01/07/2023]
Abstract
Substantial progress has been made in defining the regulatory factors involved in generating multipotent neural crest cells at the neural plate border of vertebrate embryos, controlling the onset of their migratory behavior, and directing their differentiation into one of a diverse array of derivatives. Growing evidence suggests that these factors function as a complex network, in some cases displaying overlapping functions and cross-regulatory interactions. Mechanisms are emerging for how some of these regulatory components are controlled post-translationally and the extent to which their activities are conserved across species.
Collapse
Affiliation(s)
- Kimberly M Taylor
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, IL 60208, USA
| | | |
Collapse
|
44
|
Abstract
Asymmetries in the egg, established during oogenesis, set the stage for a cascade of intercellular signaling events leading to differential gene expression and subsequent tissue and organ formation. Maternally supplied Sox-type transcription factors have recently emerged as key components in the patterning of the early embryo and the regulation of embryonic stem cell differentiation. In deuterostomes, B1-type Soxs are asymmetrically localized to the future animal/ectodermal region where they act to suppress mesendodermal, and favor neuroectodermal differentiation, while vegetally localized F-type Soxs are involved in mesendodermal differentiation. Here, we review past observations and present new data from studies on the clawed frog Xenopus laevis. Animally localized Sox3 acts to inhibit Nodal (Xnr5 and Xnr6) expression, and induces the expression of genes (Ectodermin, Xema, and Coco) whose products repress Nodal signaling. Vegetally localized Sox7 positively regulates Nodal (Xnr4, Xnr5, and Xnr6) expression, as well as the expression of genes involved in mesodermal (Xmenf, Slug, and Snail) and endodermal (Endodermin and Sox17beta) differentiation. Given the evolutionary strategy of using common regulatory networks, it seems likely that a homologous Sox-Axis is active during embryonic development in many metazoans.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Molecular, Cellular and Developmental Biology University of Colorado at Boulder Boulder, CO 80309-0347, USA
| | | |
Collapse
|
45
|
Broders-Bondon F, Chesneau A, Romero-Oliva F, Mazabraud A, Mayor R, Thiery JP. Regulation ofXSnail2 expression by Rho GTPases. Dev Dyn 2007; 236:2555-66. [PMID: 17676632 DOI: 10.1002/dvdy.21273] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We analyzed the effects of Rho GTPases on XSnail2 expression during neural crest (NC) ontogeny in Xenopus laevis embryos. The ectopic expression of both dominant-negative (N-) and constitutively active (V-) Rho GTPase mutants after RNA or DNA microinjection disrupted the endogenous expression of XSnail2, XFoxD3, and XSnail1. V14RhoA and N17Rac1 were inhibitory, whereas N19RhoA and V12Rac1 increased NC marker gene expression. In reporter assays using a XSnail2 promoter-green fluorescent protein (GFP) construct (alpha700BA-GFP), the ectopic expression of V14RhoA, N17Rac1, or the Rac1 inhibitor NSC 23766 decreased reporter expression in NC-neural plate, whereas N19RhoA or the RhoA inhibitor Y27632 and V12Rac1 enhanced it. Similarly, transgenic embryos expressing Rho GTPase mutants and GFP under control of the alpha700BA promoter displayed variations similar to those observed for ectopic RNA and DNA expression. These results show that Rho GTPases can regulate the expression of XSnail2 during NC ontogeny.
Collapse
Affiliation(s)
- Florence Broders-Bondon
- Laboratoire de Morphogenèse Cellulaire et Progression Tumorale, UMR144 CNRS / Institut Curie, Paris, France.
| | | | | | | | | | | |
Collapse
|