51
|
RNA interference targeting slug increases cholangiocarcinoma cell sensitivity to cisplatin via upregulating PUMA. Int J Mol Sci 2011; 12:385-400. [PMID: 21339993 PMCID: PMC3039959 DOI: 10.3390/ijms12010385] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 01/06/2011] [Accepted: 01/07/2011] [Indexed: 11/17/2022] Open
Abstract
Slug is an E-cadherin repressor and a suppressor of PUMA (p53 upregulated modulator of apoptosis) and it has recently been demonstrated that Slug plays an important role in controlling apoptosis. In this study, we examined whether Slug's ability to silence expression suppresses the growth of cholangiocarcinoma cells and/or sensitizes cholangiocarcinoma cells to chemotherapeutic agents through induction of apoptosis. We targeted the Slug gene using siRNA (Slug siRNA) via full Slug cDNA plasmid (Slug cDNA) transfection of cholangiocarcinoma cells. Slug siRNA, cisplatin, or Slug siRNA in combination with cisplatin, were used to treat cholangiocarcinoma cells in vitro. Western blot was used to detect the expression of Slug, PUMA, and E-cadherin protein. TUNEL, Annexin V Staining, and cell cycle analysis were used to detect apoptosis. A nude mice subcutaneous xenograft model of QBC939 cells was used to assess the effect of Slug silencing and/or cisplatin on tumor growth. Immunohistochemical staining was used to analyze the expression of Slug and PUMA. TUNEL was used to detect apoptosis in vivo. The results showed that PUMA and E-cadherin expression in cholangiocarcinoma cells is Slug dependent. We demonstrated that Slug silencing and cisplatin both promote apoptosis by upregulation of PUMA, not by upregulation of E-cadherin. Slug silencing significantly sensitized cholangiocarcinoma cells to cisplatin through upregulation of PUMA. Finally, we showed that Slug silencing suppressed the growth of QBC939 xenograft tumors and sensitized the tumor cells to cisplatin through PUMA upregulation and induction of apoptosis. Our findings indicate that Slug is an important modulator of the therapeutic response of cholangiocarcinoma cells and is potentially useful as a sensitizer in cholangiocarcinoma therapy. One of the mechanisms is the regulation of PUMA by Slug.
Collapse
|
52
|
Expression of Snail2 in long bone osteosarcomas correlates with tumour malignancy. Tumour Biol 2011; 32:515-26. [PMID: 21207222 PMCID: PMC3109975 DOI: 10.1007/s13277-010-0146-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 12/06/2010] [Indexed: 01/12/2023] Open
Abstract
Snail2 is a marker of malignancy in epithelial tumours; however, in sarcomas, it is not known if this protein is present. Here we examine the expression of Snail2 in one type of sarcoma, osteosarcoma, and explore its relationship to tumour grade, subtype and anatomical location in cases of long bone and cranial bone osteosarcoma. Long bone osteosarcomas typically have a much greater metastatic capability and a poorer prognosis. We find that Snail2 is expressed in the three main subtypes of long bone osteosarcoma—osteoblastic, chondroblastic and fibroblastic. Regression analysis showed that Snail 2 expression was statistically correlated with tumour grade (p = 0.014) in all of these subtypes. Snail2 was only expressed in high-grade cranial bone osteosarcomas, suggesting a link between Snail2 expression and metastasis. This is the first time Snail2 has been associated with any sarcoma, and this study shows that Snail2 may be a useful prognostic marker for this disease.
Collapse
|
53
|
Abstract
Cancer is a complex disease in which cells have circumvented normal restraints on tissue growth and have acquired complex abnormalities in their genomes, posing a considerable challenge to identifying the pathways and mechanisms that drive fundamental aspects of the malignant phenotype. Genetic analyses of the normal development of the nematode Caenorhabditis elegans have revealed evolutionarily conserved mechanisms through which individual cells establish their fates, and how they make and execute the decision to survive or undergo programmed cell death. The pathways identified through these studies have mammalian counterparts that are co-opted by malignant cells. Effective cancer drugs now target some of these pathways, and more are likely to be discovered.
Collapse
Affiliation(s)
- Malia B Potts
- Departments of Pediatrics and Molecular Biology, University of Texas Southwestern Medical Center at Dallas, 75390-9148, USA
| | | |
Collapse
|
54
|
Abstract
The zinc finger transcription factor Slug (Snai2) serves a wide variety of functions in the epidermis, with roles in skin development, hair growth, wound healing, skin cancer, and sunburn. Slug is expressed in basal keratinocytes and hair follicles where it is important in maintaining epidermal homeostasis. Slug also helps coordinate the skin response to exogenous stimuli. Slug is rapidly induced by a variety of growth factors and injurious agents and Slug controls, directly or indirectly, a variety of keratinocyte responses, including changes in differentiation, adhesion, motility, and production of inflammatory mediators. Slug thus modulates the interactions of the keratinocyte with its environment and with surrounding cells. The function of Slug in the epidermis appears to be distinct from that of the closely related Snail transcription factor.
Collapse
Affiliation(s)
- Stephanie H Shirley
- Department of Carcinogenesis, University of Texas MD Anderson Cancer Center, Science Park Research Division, Smithville, Texas 78957, USA
| | | | | | | |
Collapse
|
55
|
Chandler HL, Gemensky-Metzler AJ, Bras ID, Robbin-Webb TE, Saville WJA, Colitz CMH. In vivo effects of adjunctive tetracycline treatment on refractory corneal ulcers in dogs. J Am Vet Med Assoc 2010; 237:378-86. [PMID: 20707747 DOI: 10.2460/javma.237.4.378] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate effect of adjunctive treatment with tetracycline analogues on time to complete corneal reepithelialization in dogs with nonhealing (ie, refractory) corneal ulcers. DESIGN Randomized controlled clinical trial. ANIMALS 89 dogs with refractory corneal ulcers. PROCEDURES Corneal ulcers were treated via debridement and grid keratotomy. Dogs were assigned to receive 1 of 3 treatment regimens for up to 6 weeks: doxycycline (5 mg/kg [2.27 mg/lb], PO, q 12 h) with topically applied ophthalmic ointment containing neomycin, polymyxin B, and bacitracin (ie, triple antibiotic ointment; q 8 h); cephalexin (22 mg/kg [10 mg/lb], PO, q 12 h) with topically applied oxytetracycline ophthalmic ointment (q 8 h); or a control treatment of cephalexin (22 mg/kg, PO, q 12 h) with topically applied triple antibiotic ointment (q 8 h). Healing was monitored via measurements of the wound with calipers and evaluation of photographs obtained every 2 weeks. Treatment effectiveness was evaluated by wound healing and decreased signs of pain. RESULTS The Boxer breed was overrepresented in all groups. At the 2-week time point, wound healing was significantly more common in small-breed dogs, compared with large-breed dogs. Dogs treated with oxytetracycline ophthalmic ointment had a significantly shorter healing time than did dogs receiving the control treatment. Corneal ulcers in dogs that received doxycycline PO healed more rapidly than did ulcers in dogs in the control treatment group; however, this difference was not significant. CONCLUSIONS AND CLINICAL RELEVANCE Topical tetracycline ophthalmic ointment was a safe, inexpensive, and effective adjunctive treatment for refractory corneal ulcers in dogs.
Collapse
Affiliation(s)
- Heather L Chandler
- College of Optometry, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | | | |
Collapse
|
56
|
Jhingory S, Wu CY, Taneyhill L. Novel insight into the function and regulation of alphaN-catenin by Snail2 during chick neural crest cell migration. Dev Biol 2010; 344:896-910. [PMID: 20542025 PMCID: PMC2914159 DOI: 10.1016/j.ydbio.2010.06.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 06/02/2010] [Accepted: 06/02/2010] [Indexed: 12/18/2022]
Abstract
The neural crest is a transient population of migratory cells that differentiates to form a variety of cell types in the vertebrate embryo, including melanocytes, the craniofacial skeleton, and portions of the peripheral nervous system. These cells initially exist as adherent epithelial cells in the dorsal aspect of the neural tube and only later become migratory after an epithelial-to-mesenchymal transition (EMT). Snail2 plays a critical role in mediating chick neural crest cell EMT and migration due to its expression by both premigratory and migratory cranial neural crest cells and its ability to down-regulate intercellular junctions components. In an attempt to delineate the role of cellular junction components in the neural crest, we have identified the adherens junction molecule neural alpha-catenin (alphaN-catenin) as a Snail2 target gene whose repression is critical for chick neural crest cell migration. Knock-down and overexpression of alphaN-catenin enhances and inhibits neural crest cell migration, respectively. Furthermore, our results reveal that alphaN-catenin regulates the appropriate movement of neural crest cells away from the neural tube into the embryo. Collectively, our data point to a novel function of an adherens junction protein in facilitating the proper migration of neural crest cells during the development of the vertebrate embryo.
Collapse
Affiliation(s)
| | | | - L.A. Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
57
|
Das SK. Regional development of uterine decidualization: molecular signaling by Hoxa-10. Mol Reprod Dev 2010; 77:387-96. [PMID: 19921737 DOI: 10.1002/mrd.21133] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Uterine decidualization, a key event in implantation, is critically controlled by stromal cell proliferation and differentiation. Although the molecular mechanism that controls this event is not well understood, the general consensus is that the factors derived locally at the site of implantation influence aspects of decidualization. Hoxa-10, a developmentally regulated homeobox transcription factor, is highly expressed in decidualizing stromal cells, and targeted deletion of Hoxa-10 in mice shows severe decidualization defects, primarily due to the reduced stromal cell responsiveness to progesterone (P(4)). While the increased stromal cell proliferation is considered to be an initiator of decidualization, the establishment of a full-grown functional decidua appears to depend on the aspects of regional proliferation and differentiation. In this regard, this article provides an overview of potential signaling mechanisms mediated by Hoxa-10 that can influence a host of genes and cell functions necessary for propagating regional decidual development.
Collapse
Affiliation(s)
- Sanjoy K Das
- Reproductive Sciences, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229-3039, USA.
| |
Collapse
|
58
|
de Herreros AG, Peiró S, Nassour M, Savagner P. Snail family regulation and epithelial mesenchymal transitions in breast cancer progression. J Mammary Gland Biol Neoplasia 2010; 15:135-47. [PMID: 20455012 PMCID: PMC2930904 DOI: 10.1007/s10911-010-9179-8] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 04/26/2010] [Indexed: 12/12/2022] Open
Abstract
Since its initial description, the interconversion between epithelial and mesenchymal cells (designed as epithelial-mesenchymal or mesenchymal-epithelial transition, EMT or MET, respectively) has received special attention since it provides epithelial cells with migratory features. Different studies using cell lines have identified cytokines, intercellular signaling elements and transcriptional factors capable of regulating this process. Particularly, the identification of Snail family members as key effectors of EMT has opened new ways for the study of this cellular process. In this article we discuss the molecular pathways that control EMT, showing a very tight and interdependent regulation. We also analyze the contribution of EMT and Snail genes in the process of tumorigenesis using the mammary gland as cellular model.
Collapse
Affiliation(s)
- Antonio Garcia de Herreros
- IMIM-Hospital del Mar, Parc de Recerca Biomèdica de Barcelona, C/Doctor Aiguader, 88, 08003 Barcelona, Spain.
| | | | | | | |
Collapse
|
59
|
Emadi Baygi M, Soheili ZS, Essmann F, Deezagi A, Engers R, Goering W, Schulz WA. Slug/SNAI2 regulates cell proliferation and invasiveness of metastatic prostate cancer cell lines. Tumour Biol 2010; 31:297-307. [PMID: 20506051 DOI: 10.1007/s13277-010-0037-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 04/02/2010] [Indexed: 12/18/2022] Open
Abstract
Many metastatic cancers recapitulate the epithelial-to-mesenchymal transition (EMT) resulting in enhanced cell motility and invasiveness. The EMT is regulated by several transcription factors, including the zinc finger protein SNAI2, also named Slug, which appears to exert additional functions during development and cancer progression. We have studied the function of SNAI2 in prostate cancer cells. Quantitative RT-PCR analysis showed strong SNAI2 expression particularly in the PC-3 and PC3-16 prostate carcinoma cell lines. Knockdown of SNAI2 by specific siRNA induced changes in EMT markers and inhibited invasion of both cell lines into a matrigel matrix. SNAI2 siRNA-treated cells did not tolerate detachment from the culture plates, likely at least in part due to downregulation of integrin alpha6beta4. SNAI2 knockdown disturbed the microtubular and actin cytoskeletons, especially severely in PC-3 cells, resulting in grossly enlarged, flattened, and sometimes multinuclear cells. Knockdown also decreased cell proliferation, with a prominent G0/G1 arrest in PC3-16. Together, our data imply that SNAI2 exerts strong effects on the cytoskeleton and adhesion of those prostate cancer cells that express it and is necessary for their proliferation and invasiveness.
Collapse
Affiliation(s)
- Modjtaba Emadi Baygi
- Department of Genetics, Faculty of Basic Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
60
|
Lambertini E, Franceschetti T, Torreggiani E, Penolazzi L, Pastore A, Pelucchi S, Gambari R, Piva R. SLUG: a new target of lymphoid enhancer factor-1 in human osteoblasts. BMC Mol Biol 2010; 11:13. [PMID: 20128911 PMCID: PMC2834684 DOI: 10.1186/1471-2199-11-13] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 02/03/2010] [Indexed: 11/28/2022] Open
Abstract
Background Lymphoid Enhancer Factor-1 (Lef-1) is a member of a transcription factor family that acts as downstream mediator of the Wnt/β-catenin signalling pathway which plays a critical role in osteoblast proliferation and differentiation. In a search for Lef-1 responsive genes in human osteoblasts, we focused on the transcriptional regulation of the SLUG, a zinc finger transcription factor belonging to the Snail family of developmental proteins. Although the role of SLUG in epithelial-mesenchymal transition and cell motility during embryogenesis is well documented, the functions of this factor in most normal adult human tissues are largely unknown. In this study we investigated SLUG expression in normal human osteoblasts and their mesenchymal precursors, and its possible correlation with Lef-1 and Wnt/β-catenin signalling. Results The experiments were performed on normal human primary osteoblasts obtained from bone fragments, cultured in osteogenic conditions in presence of Lef-1 expression vector or GSK-3β inhibitor, SB216763. We demonstrated that the transcription factor SLUG is present in osteoblasts as well as in their mesenchymal precursors obtained from Wharton's Jelly of human umbilical cord and induced to osteoblastic differentiation. We found that SLUG is positively correlated with RUNX2 expression and deposition of mineralized matrix, and is regulated by Lef-1 and β-catenin. Consistently, Chromatin Immunoprecipitation (ChIP) assay, used to detect the direct Lef/Tcf factors that are responsible for the promoter activity of SLUG gene, demonstrated that Lef-1, TCF-1 and TCF4 are recruited to the SLUG gene promoter "in vivo". Conclusion These studies provide, for the first time, the evidence that SLUG expression is correlated with osteogenic commitment, and is positively regulated by Lef-1 signal in normal human osteoblasts. These findings will help to further understand the regulation of the human SLUG gene and reveal the biological functions of SLUG in the context of bone tissue.
Collapse
Affiliation(s)
- Elisabetta Lambertini
- Department of Biochemistry and Molecular Biology, Molecular Biology Section, University of Ferrara, Via Fossato di Mortara, 74, 44100 Ferrara, Ferrara, Italy
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Slug deficiency enhances self-renewal of hematopoietic stem cells during hematopoietic regeneration. Blood 2009; 115:1709-17. [PMID: 20032500 DOI: 10.1182/blood-2009-07-232934] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Both extrinsic and intrinsic mechanisms tightly govern hematopoietic stem cell (HSC) decisions of self-renewal and differentiation. However, transcription factors that can selectively regulate HSC self-renewal division after stress remain to be identified. Slug is an evolutionarily conserved zinc-finger transcription factor that is highly expressed in primitive hematopoietic cells and is critical for the radioprotection of these key cells. We studied the effect of Slug in the regulation of HSCs in Slug-deficient mice under normal and stress conditions using serial functional assays. Here, we show that Slug deficiency does not disturb hematopoiesis or alter HSC homeostasis and differentiation in bone marrow but increases the numbers of primitive hematopoietic cells in the extramedullary spleen site. Deletion of Slug enhances HSC repopulating potential but not its homing and differentiation ability. Furthermore, Slug deficiency increases HSC proliferation and repopulating potential in vivo after myelosuppression and accelerates HSC expansion during in vitro culture. Therefore, we propose that Slug is essential for controlling the transition of HSCs from relative quiescence under steady-state condition to rapid proliferation under stress conditions. Our data suggest that inhibition of Slug in HSCs may present a novel strategy for accelerating hematopoietic recovery, thus providing therapeutic benefits for patients after clinical myelosuppressive treatment.
Collapse
|
62
|
MacPherson MR, Molina P, Souchelnytskyi S, Wernstedt C, Martin-Pérez J, Portillo F, Cano A. Phosphorylation of serine 11 and serine 92 as new positive regulators of human Snail1 function: potential involvement of casein kinase-2 and the cAMP-activated kinase protein kinase A. Mol Biol Cell 2009; 21:244-53. [PMID: 19923321 PMCID: PMC2808231 DOI: 10.1091/mbc.e09-06-0504] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
New phosphorylation sites in Snail1 have been identified with functional implications. Serines 11 and 92 participate in the control of Snail1 stability and positively regulate Snail1 repressive function and its interaction with mSin3A co-repressor. Furthermore, serines 11 and 92 are required for Snail1-mediated EMT and cell viability, respectively. Snail1 is a major factor for epithelial-mesenchymal transition (EMT), an important event in tumor metastasis and in other pathologies. Snail1 is tightly regulated at transcriptional and posttranscriptional levels. Control of Snail1 protein stability and nuclear export by GSK3β phosphorylation is important for Snail1 functionality. Stabilization mechanisms independent of GSK3β have also been reported, including interaction with LOXL2 or regulation of the COP9 signalosome by inflammatory signals. To get further insights into the role of Snail1 phosphorylation, we have performed an in-depth analysis of in vivo human Snail1 phosphorylation combined with mutational studies. We identify new phosphorylation sites at serines 11, 82, and 92 and confirmed previously suggested phosphorylations at serine 104 and 107. Serines 11 and 92 participate in the control of Snail1 stability and positively regulate Snail1 repressive function and its interaction with mSin3A corepressor. Furthermore, serines 11 and 92 are required for Snail1-mediated EMT and cell viability, respectively. PKA and CK2 have been characterized as the main kinases responsible for in vitro Snail1 phosphorylation at serine 11 and 92, respectively. These results highlight serines 11 and 92 as new players in Snail1 regulation and suggest the participation of CK2 and PKA in the modulation of Snail1 functionality.
Collapse
Affiliation(s)
- Matthew Reid MacPherson
- Departamento de Bioquímica, UAM, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, 28029 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
63
|
Reece-Hoyes JS, Deplancke B, Barrasa MI, Hatzold J, Smit RB, Arda HE, Pope PA, Gaudet J, Conradt B, Walhout AJM. The C. elegans Snail homolog CES-1 can activate gene expression in vivo and share targets with bHLH transcription factors. Nucleic Acids Res 2009; 37:3689-98. [PMID: 19372275 PMCID: PMC2699517 DOI: 10.1093/nar/gkp232] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Snail-type transcription factors (TFs) are found in numerous metazoan organisms and function in a plethora of cellular and developmental processes including mesoderm and neuronal development, apoptosis and cancer. So far, Snail-type TFs are exclusively known as transcriptional repressors. They repress gene expression by recruiting transcriptional co-repressors and/or by preventing DNA binding of activators from the basic helix-loop-helix (bHLH) family of TFs to CAGGTG E-box sequences. Here we report that the Caenorhabditis elegans Snail-type TF CES-1 can activate transcription in vivo. Moreover, we provide results that suggest that CES-1 can share its binding site with bHLH TFs, in different tissues, rather than only occluding bHLH DNA binding. Together, our data indicate that there are at least two types of CES-1 target genes and, therefore, that the molecular function of Snail-type TFs is more plastic than previously appreciated.
Collapse
Affiliation(s)
- John S Reece-Hoyes
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Xu J, Lamouille S, Derynck R. TGF-beta-induced epithelial to mesenchymal transition. Cell Res 2009; 19:156-72. [PMID: 19153598 DOI: 10.1038/cr.2009.5] [Citation(s) in RCA: 2149] [Impact Index Per Article: 134.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During development and in the context of different morphogenetic events, epithelial cells undergo a process called epithelial to mesenchymal transition or transdifferentiation (EMT). In this process, the cells lose their epithelial characteristics, including their polarity and specialized cell-cell contacts, and acquire a migratory behavior, allowing them to move away from their epithelial cell community and to integrate into surrounding tissue, even at remote locations. EMT illustrates the differentiation plasticity during development and is complemented by another process, called mesenchymal to epithelial transition (MET). While being an integral process during development, EMT is also recapitulated under pathological conditions, prominently in fibrosis and in invasion and metastasis of carcinomas. Accordingly, EMT is considered as an important step in tumor progression. TGF-beta signaling has been shown to play an important role in EMT. In fact, adding TGF-beta to epithelial cells in culture is a convenient way to induce EMT in various epithelial cells. Although much less characterized, epithelial plasticity can also be regulated by TGF-beta-related bone morphogenetic proteins (BMPs), and BMPs have been shown to induce EMT or MET depending on the developmental context. In this review, we will discuss the induction of EMT in response to TGF-beta, and focus on the underlying signaling and transcription mechanisms.
Collapse
Affiliation(s)
- Jian Xu
- Department of Cell and Tissue Biology, Programs in Cell Biology and Developmental Biology, University of California-San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
65
|
Arnoux V, Nassour M, L'Helgoualc'h A, Hipskind RA, Savagner P. Erk5 controls Slug expression and keratinocyte activation during wound healing. Mol Biol Cell 2008; 19:4738-49. [PMID: 18716062 DOI: 10.1091/mbc.e07-10-1078] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Reepithelialization during cutaneous wound healing involves numerous signals that result in basal keratinocyte activation, spreading, and migration, all linked to a loosening of cell-cell adhesion structures. The transcription factor Slug is required for this process, and EGF treatment of human keratinocytes induced activating phosphorylation of Erk5 that coincides with slug transcription. Accordingly, ectopic activation of Erk5 led to increased Slug mRNA levels and faster wound healing, whereas keratinocyte migration was totally blocked by Erk5 pathway inhibition. Expression of a shRNA specific for Erk5 strongly diminished Erk5 levels in keratinocytes and significantly decreased their motility response to EGF, along with induction of Slug expression. These Erk5-deprived keratinocytes showed an altered, more compact morphology, along with disruption of desmosome organization. Accordingly, they displayed an altered ability to form cell aggregates. These results implicate a novel EGFR/Erk5/Slug pathway in the control of cytoskeleton organization and cell motility in keratinocytes treated with EGF.
Collapse
Affiliation(s)
- Valerie Arnoux
- INSERM EMI 229, Genotypes et phenotypes tumoraux, Centre de Recherche en Cancerologie de Montpellier, CRLC Val d'Aurelle-Paul Lamarque, 34298 Montpellier, France
| | | | | | | | | |
Collapse
|
66
|
Abstract
We previously reported ultraviolet radiation (UVR) induction of Slug, a Snail family zinc-finger transcription factor, in the epidermis of mice; we now report that Slug-knockout mice are, unexpectedly, more resistant to sunburn than wild-type mice. There was a marked difference between the cutaneous inflammatory response in the skin of Slug-knockout and wild-type mice from 12 h to 1 week following a single exposure to 3 minimal erythemal doses of UVR. Slug-knockout mice showed a much reduced immediate increase in skin thickness and neutrophil infiltration compared to wild-type mice. However, there were as many or more intraepidermal T cells, dermal mast cells, and dermal blood vessels in the UVR-exposed skin of Slug-knockout mice as in the skin of wild-type mice. Differences in cytokine and chemokine expression following UVR appeared to account for at least some differences between the genotypes in cutaneous inflammatory response. Despite the reported antiapoptotic and antiproliferative role for Slug in some cell types, we observed little difference between the genotypes in UVR-induced keratinocyte apoptosis or proliferation. Our findings indicate an unexpected but important role for Slug in the acute cutaneous inflammatory response to UVR.
Collapse
|
67
|
Mittal MK, Myers JN, Misra S, Bailey CK, Chaudhuri G. In vivo binding to and functional repression of the VDR gene promoter by SLUG in human breast cells. Biochem Biophys Res Commun 2008; 372:30-4. [PMID: 18485278 PMCID: PMC2846599 DOI: 10.1016/j.bbrc.2008.04.187] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 04/28/2008] [Indexed: 10/22/2022]
Abstract
The regulation of vitamin D receptor (VDR), a key mediator in the vitamin D pathway, in breast cancer etiology has long been of interest. We have shown here that the transcriptional repressor protein SLUG inhibits the expression of VDR in human breast cancer cells. To explore the possibility that SLUG regulates the VDR gene promoter, we cloned a 628bp fragment (-613 to +15) of the human VDR gene promoter. This region contains three E2-box sequences (CAGGTG/CACCTG), the classical binding site of SLUG. SLUG specifically inhibited VDR gene promoter activity. Chromatin-immunoprecipitation (ChIP) assays revealed that SLUG is recruited on the native VDR gene promoter along with the co-repressor protein CtBP1 and the effector protein HDAC1. These data suggests that SLUG binds to the E2-box sequences of the VDR gene promoter and recruits CtBP1 and HDAC1, which results in the inhibition of VDR gene expression by chromatin remodeling.
Collapse
Affiliation(s)
- Mukul K. Mittal
- Department of Microbial Pathogenesis & Immune Response, Meharry Medical College, 1005 D.B. Todd Jr. Boulevard, Nashville, TN 37208, USA
| | - Jeremy N. Myers
- Department of Microbial Pathogenesis & Immune Response, Meharry Medical College, 1005 D.B. Todd Jr. Boulevard, Nashville, TN 37208, USA
| | - Smita Misra
- Department of Microbial Pathogenesis & Immune Response, Meharry Medical College, 1005 D.B. Todd Jr. Boulevard, Nashville, TN 37208, USA
| | - Charvann K. Bailey
- Department of Microbial Pathogenesis & Immune Response, Meharry Medical College, 1005 D.B. Todd Jr. Boulevard, Nashville, TN 37208, USA
| | - Gautam Chaudhuri
- Department of Microbial Pathogenesis & Immune Response, Meharry Medical College, 1005 D.B. Todd Jr. Boulevard, Nashville, TN 37208, USA
| |
Collapse
|
68
|
Chandler HL, Kusewitt DF, Colitz CMH. Modulation of matrix metalloproteinases by ultraviolet radiation in the canine cornea. Vet Ophthalmol 2008; 11:135-44. [DOI: 10.1111/j.1463-5224.2008.00575.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
69
|
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 PMCID: PMC2279146 DOI: 10.1016/j.devcel.2008.01.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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 and Cell Biology, Washington University, Saint Louis, MO
| | - Yunfeng Feng
- Department of Medicine and Cell Biology, Washington University, Saint Louis, MO
| | | | | | - Kristen L. Kroll
- Department of Molecular Biology and Pharmacology, Washington University, Saint Louis, MO
| | - Gregory D. Longmore
- Department of Medicine and Cell Biology, Washington University, Saint Louis, MO
| |
Collapse
|
70
|
Cobaleda C, Pérez-Caro M, Vicente-Dueñas C, Sánchez-García I. Function of the Zinc-Finger Transcription FactorSNAI2in Cancer and Development. Annu Rev Genet 2007; 41:41-61. [PMID: 17550342 DOI: 10.1146/annurev.genet.41.110306.130146] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Elucidation of the molecular mechanisms that underlie disease development is still a tremendous challenge for basic science, and a prerequisite to the development of new and disease-specific targeted therapies. This review focuses on the function of SNAI2, a member of the Snail family of zinc-finger transcription factors, and discusses its possible role in disease development. SNAI2 has been implicated in diseases of melanocyte development and cancer in humans. Many malignancies arise from a rare population of cells that alone have the ability to self-renew and sustain the tumor (i.e., cancer stem cells). SNAI2 controls key aspects of stem cell function in mouse and human, suggesting that similar mechanisms control normal development and cancer stem cell properties. These insights are expected to contribute significantly to the genetics of cancer and to the development of both cancer therapy and new methods for assessing treatment efficacy.
Collapse
Affiliation(s)
- César Cobaleda
- Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | | | | | | |
Collapse
|
71
|
Ayyanathan K, Peng H, Hou Z, Fredericks WJ, Goyal RK, Langer EM, Longmore GD, Rauscher FJ. The Ajuba LIM domain protein is a corepressor for SNAG domain mediated repression and participates in nucleocytoplasmic Shuttling. Cancer Res 2007; 67:9097-106. [PMID: 17909014 DOI: 10.1158/0008-5472.can-07-2987] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The SNAG repression domain is comprised of a highly conserved 21-amino acid sequence, is named for its presence in the Snail/growth factor independence-1 class of zinc finger transcription factors, and is present in a variety of proto-oncogenic transcription factors and developmental regulators. The prototype SNAG domain containing oncogene, growth factor independence-1, is responsible for the development of T cell thymomas. The SNAIL proteins also encode the SNAG domain and play key roles in epithelial mesenchymal differentiation events during development and metastasis. Significantly, these oncogenic functions require a functional SNAG domain. The molecular mechanisms of SNAG domain-mediated transcriptional repression are largely unknown. Using a yeast two-hybrid strategy, we identified Ajuba, a multiple LIM domain protein that can function as a corepressor for the SNAG domain. Ajuba interacts with the SNAG domain in vitro and in vivo, colocalizes with it, and enhances SNAG-mediated transcriptional repression. Ajuba shuttles between the cytoplasm and the nucleus and may form a novel intracellular signaling system. Using an integrated reporter gene combined with chromatin immunoprecipitation, we observed rapid, SNAG-dependent assembly of a multiprotein complex that included Ajuba, SNAG, and histone modifications consistent with the repressed state. Thus, SNAG domain proteins may bind Ajuba, trapping it in the nucleus where it functions as an adapter or molecular scaffold for the assembly of macromolecular repression complexes at target promoters.
Collapse
|
72
|
Kim HS, Kim MS, Hancock AL, Harper JCP, Park JY, Poy G, Perantoni AO, Cam M, Malik K, Lee SB. Identification of Novel Wilms' Tumor Suppressor Gene Target Genes Implicated in Kidney Development. J Biol Chem 2007; 282:16278-87. [PMID: 17430890 DOI: 10.1074/jbc.m700215200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Wilms' tumor suppressor gene (WT1) encodes a zinc finger transcription factor that is vital during development of several organs including metanephric kidneys. Despite the critical regulatory role of WT1, the pathways and mechanisms by which WT1 orchestrates development remain elusive. To identify WT1 target genes, we performed a genome-wide expression profiling analysis in cells expressing inducible WT1. We identified a number of direct WT1 target genes, including the epidermal growth factor (EGF)-family ligands epiregulin and HB-EGF, the chemokine CX3CL1, and the transcription factors SLUG and JUNB. The target genes were validated using quantitative reverse transcriptase-polymerase chain reaction, small interfering RNA knockdowns, chromatin immunoprecipitation, and luciferase reporter analyses. Immunohistochemistry of fetal kidneys confirmed that a number of the WT1 target genes had overlapping expression patterns with the highly restricted spatiotemporal expression of WT1. Finally, using an in vitro embryonic kidney culture assay, we found that the addition of recombinant epiregulin, amphiregulin, CX3CL1, and interleukin-11 significantly enhanced ureteric bud branching morphogenesis. Our genome-wide screen implicates WT1 in the transcriptional regulation of the EGF-family of growth factors as well as the CX3CL1 chemokine during nephrogenesis.
Collapse
Affiliation(s)
- Ho-Shik Kim
- Genetics of Development and Disease Branch, Microarray Core Facility, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 2007; 7:415-28. [PMID: 17508028 DOI: 10.1038/nrc2131] [Citation(s) in RCA: 2488] [Impact Index Per Article: 138.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The molecular mechanisms that underlie tumour progression are still poorly understood, but recently our knowledge of particular aspects of some of these processes has increased. Specifically, the identification of Snail, ZEB and some basic helix-loop-helix (bHLH) factors as inducers of epithelial-mesenchymal transition (EMT) and potent repressors of E-cadherin expression has opened new avenues of research with potential clinical implications.
Collapse
Affiliation(s)
- Héctor Peinado
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | | | | |
Collapse
|
74
|
Alves CC, Rosivatz E, Schott C, Hollweck R, Becker I, Sarbia M, Carneiro F, Becker KF. Slug is overexpressed in gastric carcinomas and may act synergistically with SIP1 and Snail in the down-regulation of E-cadherin. J Pathol 2007; 211:507-515. [PMID: 17299729 DOI: 10.1002/path.2138] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Epithelial-mesenchymal transition (EMT) involving down-regulation of E-cadherin is known to play an important role in tumour progression. The aim of our study was to investigate the mRNA expression of two EMT regulators-Slug and E12/E47-in primary human gastric carcinomas and to compare this with the expression of E-cadherin and other EMT regulators (Snail, Twist, and SIP1). We studied a series of 59 gastric carcinomas by real-time quantitative RT-PCR in formalin-fixed and paraffin-embedded tissues. Thirty-four cases (58%) showed Slug up-regulation in the tumour; reduced or negative expression of E-cadherin was present in 24 of these (71%, p<0.0001). Twenty-one cases (36%) showed E12/E47 up-regulation that was not significantly associated with E-cadherin down-regulation (p=0.5734). Slug up-regulation accompanied by E-cadherin down-regulation correlated with the presence of distant metastases (p=0.0029) and with advanced pTNM stages (p=0.0424). A statistically significant association was found between Slug up-regulation and the expression of SIP1 in intestinal (p=0.0014) and Snail in diffuse (p=0.0067) carcinomas. We present the first study integrating the analysis of several EMT regulators in primary gastric carcinomas and conclude that Slug up-regulation is associated with E-cadherin down-regulation in diffuse and intestinal-type gastric carcinoma, and that this effect could be complemented by the presence of other EMT regulators.
Collapse
Affiliation(s)
- C Castro Alves
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Rua Dr Roberto Frias, S/N 4200-465 Porto, Portugal
| | - E Rosivatz
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
- Division of Cell and Molecular Biology, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - C Schott
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
| | - R Hollweck
- Institut für Medizinische Statistik und Epidemiologie, Technische Universität München, Ismaningerstrasse 22, D-81675 Munich, Germany
| | - I Becker
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
- Pathologie Rosenheim, Germany
| | - M Sarbia
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
- Institut für Pathologie, Krankenhaus Lichtenberg, Berlin, Germany
| | - F Carneiro
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Rua Dr Roberto Frias, S/N 4200-465 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto and Hospital de S João, Alameda Hernâni Monteiro, 4200-319 Porto, Portugal
| | - K-F Becker
- Institut für Pathologie, Technische Universität München, Trogerstrasse 18, D-81765 Munich, Germany
| |
Collapse
|
75
|
Taneyhill LA, Coles EG, Bronner-Fraser M. Snail2 directly represses cadherin6B during epithelial-to-mesenchymal transitions of the neural crest. Development 2007; 134:1481-90. [PMID: 17344227 PMCID: PMC2595139 DOI: 10.1242/dev.02834] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The neural crest, a transient population of migratory cells, forms the craniofacial skeleton and peripheral nervous system, among other derivatives in vertebrate embryos. The transcriptional repressor Snail2 is thought to be crucial for the epithelial-to-mesenchymal transition (EMT) that promotes neural crest delamination from the neural tube; however, little is known about its downstream targets. To this end, we depleted avian Snail2 in the premigratory neural crest using morpholino antisense oligonucleotides and examined effects on potential targets by quantitative PCR. Several dorsal neural tube genes were upregulated by alleviating Snail2 repression; moreover, the cell adhesion molecule cadherin6B was derepressed within 30 minutes of blocking Snail2 translation. Examination of the chick cadherin6B genomic sequence reveals that the regulatory region contains three pairs of clustered E boxes, representing putative Snail2 binding sites. Furthermore, in vivo and in vitro biochemical analyses demonstrate that Snail2 directly binds to these sites and regulates cadherin6B transcription. These results are the first to describe a direct target of Snail2 repression in vivo and in the context of the EMT that characterizes neural crest development.
Collapse
|
76
|
Chandler HL, Colitz CMH, Lu P, Saville WJA, Kusewitt DF. The role of the slug transcription factor in cell migration during corneal re-epithelialization in the dog. Exp Eye Res 2007; 84:400-11. [PMID: 17196588 DOI: 10.1016/j.exer.2006.10.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Revised: 09/07/2006] [Accepted: 10/16/2006] [Indexed: 01/20/2023]
Abstract
Epithelial cell migration during corneal wound re-epithelialization shares features with the developmental process of epithelial-mesenchymal transition (EMT) modulated by Snail family transcription factors, including Slug. Our studies demonstrated that Slug expression was enhanced at sites of epithelial cell migration at the margins of normally healing corneal wounds in dogs, but significantly decreased at the margins of non-healing canine corneal erosions. Increased Slug expression was associated with internalization of E-cadherin and beta-catenin from the cell membrane and with enhanced expression of smooth-muscle-specific alpha-actin, tropomyosin, and matrix metalloproteinases at wound margins. Enhanced Slug expression in corneal explants due to an adenoviral expression construct or to oxytetracycline treatment resulted in significantly higher rates of corneal epithelial cell migration. Oxytetracycline appeared to act by stimulating transforming growth factor-beta activity, thus increasing Slug expression and enhancing corneal epithelial migration. These findings highlight the similarities between epithelial migration during EMT and during successful corneal wound healing, support an important role for the Snail family in the process, and indicate that modulating Slug expression may be clinically useful in treating non-healing corneal wounds.
Collapse
Affiliation(s)
- Heather L Chandler
- Veterinary Biosciences, The Ohio State University, 1900 Coffey Road, Columbus, OH 43210, USA.
| | | | | | | | | |
Collapse
|
77
|
Bailey CK, Misra S, Mittal MK, Chaudhuri G. Human SLUG does not directly bind to CtBP1. Biochem Biophys Res Commun 2007; 353:661-4. [PMID: 17194444 PMCID: PMC3085993 DOI: 10.1016/j.bbrc.2006.12.097] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Accepted: 12/09/2006] [Indexed: 11/17/2022]
Abstract
SLUG is a transcriptional repressor protein implicated to have major role in the oncogenesis and metastasis of human breast cells. We previously have shown by chromatin immunoprecipitation assay that human SLUG (hSLUG) is co-localized with the co-repressor protein CtBP1 as bound to the BRCA2 gene silencer [M.K. Tripathi, S. Misra, S.V. Khedkar, N. Hamilton, C. Irvin-Wilson,, C. Sharan, L. Sealy, G. Chaudhuri, J. Biol. Chem. 280 (2005) 17163-17171]. hSLUG was predicted to be binding directly to CtBP1 because of an apparent presence of CtBP1 binding site in its amino acid sequences. Here, we provide evidence through yeast two-hybrid and in vitro co-immunoprecipitation analyses that hSLUG does not directly interacts with hCtBP1. This observation will help in the study of the mode of action of hSLUG in human cells.
Collapse
Affiliation(s)
| | | | - Mukul K. Mittal
- Division of Cancer Biology, Department of Biomedical Sciences, Meharry Medical College, Nashville, TN 37208, USA
| | - Gautam Chaudhuri
- Division of Cancer Biology, Department of Biomedical Sciences, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
78
|
Hayashida Y, Urata Y, Muroi E, Kono T, Miyata Y, Nomata K, Kanetake H, Kondo T, Ihara Y. Calreticulin Represses E-cadherin Gene Expression in Madin-Darby Canine Kidney Cells via Slug. J Biol Chem 2006; 281:32469-84. [PMID: 16943193 DOI: 10.1074/jbc.m607240200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Calreticulin (CRT) is a multifunctional Ca(2+)-binding molecular chaperone in the endoplasmic reticulum. In mammals, the expression level of CRT differs markedly in a variety of organs and tissues, suggesting that CRT plays a specific role in each cell type. In the present study, we focused on CRT functions in the kidney, where overall expression of CRT is quite low, and established CRT-overexpressing kidney epithelial cell-derived Madin-Darby canine kidney cells by gene transfection. We demonstrated that, in CRT-overexpressing cells, the morphology was apparently changed, and the original polarized epithelial cell phenotype was destroyed. Furthermore, CRT-overexpressing cells showed enhanced migration through Matrigel-coated Boyden chamber wells, compared with controls. E-cadherin expression was significantly suppressed at the protein and transcriptional levels in CRT-overexpressing cells compared with controls. On the other hand, the expression of mesenchymal protein markers, such as N-cadherin and fibronectin, was up-regulated. We also found that the expression of Slug, a repressor of the E-cadherin promoter, was up-regulated by overexpression of CRT through altered Ca(2+) homeostasis, and this led to enhanced binding of Slug to the E-box element in the E-cadherin promoter. Thus, we conclude that CRT regulates the epithelial-mesenchymal transition-like change of cellular phenotype by modulating the Slug/E-cadherin pathway through altered Ca(2+) homeostasis in cells, suggesting a novel function of CRT in cell-cell interaction of epithelial cells.
Collapse
Affiliation(s)
- Yasushi Hayashida
- Department of Biochemistry and Molecular Biology in Disease, Atomic Bomb Disease Institute, and Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Turner FE, Broad S, Khanim FL, Jeanes A, Talma S, Hughes S, Tselepis C, Hotchin NA. Slug regulates integrin expression and cell proliferation in human epidermal keratinocytes. J Biol Chem 2006; 281:21321-21331. [PMID: 16707493 DOI: 10.1074/jbc.m509731200] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The human epidermis is a self-renewing epithelial tissue composed of several layers of keratinocytes. Within the epidermis there exists a complex array of cell adhesion structures, and many of the cellular events within the epidermis (differentiation, proliferation, and migration) require that these adhesion structures be remodeled. The link between cell adhesion, proliferation, and differentiation within the epidermis is well established, and in particular, there is strong evidence to link the process of terminal differentiation to integrin adhesion molecule expression and function. In this paper, we have analyzed the role of a transcriptional repressor called Slug in the regulation of adhesion molecule expression and function in epidermal keratinocytes. We report that activation of Slug, which is expressed predominantly in the basal layer of the epidermis, results in down-regulation of a number of cell adhesion molecules, including E-cadherin, and several integrins, including alpha3, beta1, and beta4. We demonstrate that Slug binds to the alpha3 promoter and that repression of alpha3 transcription by Slug is dependent on an E-box sequence within the promoter. This reduction in integrin expression is reflected in decreased cell adhesion to fibronectin and laminin-5. Despite the reduction in integrin expression and function, we do not observe any increase in differentiation. We do, however, find that activation of Slug results in a significant reduction in keratinocyte proliferation.
Collapse
Affiliation(s)
- Frances E Turner
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Simon Broad
- Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | - Farhat L Khanim
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Alexa Jeanes
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Sonia Talma
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Sharon Hughes
- School of Medicine, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Chris Tselepis
- School of Medicine, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Neil A Hotchin
- School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom.
| |
Collapse
|
80
|
Saito T, Nagai M, Ladanyi M. SYT-SSX1 and SYT-SSX2 Interfere with Repression of E-Cadherin by Snail and Slug: A Potential Mechanism for Aberrant Mesenchymal to Epithelial Transition in Human Synovial Sarcoma. Cancer Res 2006; 66:6919-27. [PMID: 16849535 DOI: 10.1158/0008-5472.can-05-3697] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Synovial sarcoma is a primitive mesenchymal neoplasm characterized in almost all cases by a t(X;18) fusing the SYT transcriptional coactivator gene with either SSX1 or SSX2, with the resulting fusion gene encoding an aberrant transcriptional regulator. A subset of synovial sarcoma, predominantly cases with the SYT-SSX1 fusion, shows foci of morphologic epithelial differentiation in the form of nests of glandular epithelium. The striking spontaneous mesenchymal to epithelial differentiation in this cancer is reminiscent of a developmental switch, but the only clue to its mechanistic basis has been the observation that most cases of synovial sarcoma with glandular epithelial differentiation (GED) contain SYT-SSX1 instead of SYT-SSX2. We report here that SYT-SSX1 and SYT-SSX2 interact preferentially with Snail or Slug, respectively, and prevent these transcriptional repressors from binding to the proximal E-cadherin promoter as shown by coimmunoprecipitation and chromatin immunoprecipitation. Luciferase reporter assays reveal that SYT-SSX1 and SYT-SSX2 can respectively overcome the Snail- or Slug-mediated repression of E-cadherin transcription. This provides a mechanism by which E-cadherin expression, a prerequisite of epithelial differentiation, is aberrantly derepressed in synovial sarcoma and may also explain the association of GED with the SYT-SSX1 fusion because it interferes with Snail, the stronger repressor of the E-cadherin promoter. Thus, our data provide a mechanistic basis for the observed heterogeneity in the acquisition of epithelial characteristics in synovial sarcoma and highlight the potential role of differential interactions with Snail or Slug in modulating this phenotypic transition.
Collapse
Affiliation(s)
- Tsuyoshi Saito
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | |
Collapse
|
81
|
Sakai D, Suzuki T, Osumi N, Wakamatsu Y. Cooperative action of Sox9, Snail2 and PKA signaling in early neural crest development. Development 2006; 133:1323-33. [PMID: 16510505 DOI: 10.1242/dev.02297] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In neural crest formation, transcription factors, such as group E Sox and Snail1/Snail2 (Slug) regulate subsequent epithelial-mesenchymal transition (EMT) and migration. In particular, Sox9 has a strong effect on neural crest formation, EMT and differentiation of crest-derived cartilages in the cranium. It remains unclear, however, how Sox9 functions in these events, and how Sox9 activity is regulated. In this study, our gain-of-function and loss-of-function experiments reveal that Sox9 is essential for BMP signal-mediated induction of Snail2 and subsequent EMT in avian neural crest. We also show that Snail2 activates the Snail2 promoter, although Snail family proteins have been known as a repressor. Consistently, Sox9 directly activates the Snail2 promoter in synergy with, and through a direct binding to,Snail2. Finally, functions of these transcription factors in neural crest cells are enhanced by PKA signaling.
Collapse
Affiliation(s)
- Daisuke Sakai
- Center for Translational and Advanced Animal Research on Human Diseases, Division of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | | | | | | |
Collapse
|
82
|
Wu WS, Heinrichs S, Xu D, Garrison SP, Zambetti GP, Adams JM, Look AT. Slug antagonizes p53-mediated apoptosis of hematopoietic progenitors by repressing puma. Cell 2006; 123:641-53. [PMID: 16286009 DOI: 10.1016/j.cell.2005.09.029] [Citation(s) in RCA: 322] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2004] [Revised: 06/16/2005] [Accepted: 09/30/2005] [Indexed: 11/17/2022]
Abstract
In response to DNA damage, the p53 tumor suppressor can elicit either apoptosis or cell-cycle arrest and repair, but how this critical decision is made in specific cell types remains largely undefined. We investigated the mechanism by which the transcriptional repressor Slug specifically rescues hematopoietic progenitor cells from lethal doses of gamma radiation. We show that Slug is transcriptionally induced by p53 upon irradiation and then protects the damaged cell from apoptosis by directly repressing p53-mediated transcription of puma, a key BH3-only antagonist of the antiapoptotic Bcl-2 proteins. We established the physiologic significance of Slug-mediated repression of puma by demonstrating that mice deficient in both genes survive doses of total-body irradiation that lethally deplete hematopoietic progenitor populations in mice lacking only slug. Thus, Slug functions downstream of p53 in developing blood cells as a critical switch that prevents their apoptosis by antagonizing the trans-activation of puma by p53.
Collapse
Affiliation(s)
- Wen-Shu Wu
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
83
|
Liu YN, Lee WW, Wang CY, Chao TH, Chen Y, Chen JH. Regulatory mechanisms controlling human E-cadherin gene expression. Oncogene 2006; 24:8277-90. [PMID: 16116478 DOI: 10.1038/sj.onc.1208991] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In cancer cells, loss of E-cadherin gene expression caused dysfunction of the cell-cell junction system, triggering cancer invasion and metastasis. Therefore, E-cadherin is an important tumor-suppressor gene. To understand how E-cadherin gene expression is regulated in cancer cells, we have used E-cadherin-positive and -negative expressing cells to find out the possible up- or down regulating transcription factors in human E-cadherin regulatory sequences. Functional analysis of human E-cadherin regulatory sequences constructs indicated that AML1, Sp1, and p300 may play important roles in promoting E-cadherin expression. In addition, we found there are four HNF3-binding sites in human E-cadherin regulatory sequences. The exogenous HNF3 can enhance the E-cadherin promoter activity in metastatic breast cancer cells and the metastatic breast cancer cells stably transfected with HNF3 showed re-expression of E-cadherin. The HNF3 stable transfectants changed from mesenchymal-like into epithelial morphology. The transwell assays showed the re-expressed E-cadherin reduced cell motility of metastatic breast cancer cells. These results suggested HNF3 may play important roles in the upregulation of the E-cadherin promoter, with the consequent re-expression of E-cadherin, thus reducing the metastatic potential of breast cancer cells. These findings suggested HNF3 plays important roles in the upregulation of the E-cadherin gene and may be able to reduce the motility of metastatic breast cancer cells.
Collapse
Affiliation(s)
- Yan-Nan Liu
- Graduate Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | | | | | | | | | | |
Collapse
|
84
|
De Craene B, van Roy F, Berx G. Unraveling signalling cascades for the Snail family of transcription factors. Cell Signal 2005; 17:535-47. [PMID: 15683729 DOI: 10.1016/j.cellsig.2004.10.011] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2004] [Accepted: 10/08/2004] [Indexed: 10/26/2022]
Abstract
During development and carcinogenesis, the gradient of different molecular factors, the availability of corresponding receptors and the interplay between different signalling cascades combine to orchestrate the different stages. A good understanding of both developmental processes and oncogenesis leads to new insights into normal and aberrant regulation, processes that share some mutual key players. In this review, we will focus on the Snail family of transcription factors. These proteins, which share an evolutionarily conserved role in invertebrates and vertebrates, are implicated in several developmental processes, but are involved in carcinogenesis as well. We will highlight the different signalling cascades leading to the expression of Snail and Slug and how these factors are regulated on the transcriptional level. Then we will focus on how these factors execute their functions by repression of the numerous target genes that have been described to date.
Collapse
Affiliation(s)
- Bram De Craene
- Unit of Molecular and Cellular Oncology, Department for Molecular Biomedical Research, VIB-Ghent University, Technologiepark 927, B-9052 Ghent (Zwijnaarde), Belgium
| | | | | |
Collapse
|
85
|
Abstract
Numerous studies suggest that loss of E-cadherin is necessary to induce Epithelial–mesenchymal transition (EMT) and metastasis. Snail is a major contributor to EMTs. The Snail family of zinc-finger transcription factors interact with the E-cadherin promoter to repress transcription during EMT. The present article reviews the regulation of E-cadherin and discusses recent novel insights into the molecular basis in the process of EMT.
Collapse
|
86
|
Takahashi E, Funato N, Higashihori N, Hata Y, Gridley T, Nakamura M. Snail regulates p21(WAF/CIP1) expression in cooperation with E2A and Twist. Biochem Biophys Res Commun 2005; 325:1136-44. [PMID: 15555546 DOI: 10.1016/j.bbrc.2004.10.148] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2004] [Indexed: 01/15/2023]
Abstract
Snail, a zinc-finger transcriptional repressor, is essential for mesoderm and neural crest cell formation and epithelial-mesenchymal transition. The basic helix-loop-helix transcription factors E2A and Twist have been linked with Snail during embryonic development. In this study, we examined the role of Snail in cellular differentiation through regulation of p21(WAF/CIP1) expression. A reporter assay with the p21 promoter demonstrated that Snail inhibited expression of p21 induced by E2A. Co-expression of Snail with Twist showed additive inhibitory effects. Deletion mutants of the p21 promoter revealed that sequences between -270 and -264, which formed a complex with unidentified nuclear factor(s), were critical for E2A and Snail function. The E2A-dependent expression of the endogenous p21 gene was also inhibited by Snail.
Collapse
Affiliation(s)
- Eishi Takahashi
- Human Gene Sciences Center, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | | | | | | | | | | |
Collapse
|
87
|
Parent AE, Choi C, Caudy K, Gridley T, Kusewitt DF. The developmental transcription factor slug is widely expressed in tissues of adult mice. J Histochem Cytochem 2004; 52:959-65. [PMID: 15208362 DOI: 10.1369/jhc.4a6277.2004] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The Slug transcription factor plays an important role in epithelial-mesenchymal transformation during embryogenesis and is expressed in adult tissues during carcinogenesis. By detecting expression of a Slug-beta-galactosidase fusion protein, we have now demonstrated that Slug is also re-expressed in a variety of normal tissues in the adult mouse. Slug is expressed at relatively high levels in patchy stretches of basal cells in stratified and pseudostratified epithelium, including skin, oral mucosa, esophagus, stomach, rectum, cervix, and trachea. Slug is also found at variable levels in fibroblasts and stromal smooth muscle cells in many tissues. Sites of more intense Slug expression in mesenchymal tissues include cartilage, kidney glomeruli, lung, ovary, and uterus. Therefore, Slug expression is not restricted to the period of embryonic development or to pathological processes. The pattern of localization to basal cells in various epithelia suggests that Slug may play a role in the cell migration that occurs during continual renewal of these tissues.
Collapse
Affiliation(s)
- Allison E Parent
- Department of Veterinary Biosciences, Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
88
|
Oloumi A, McPhee T, Dedhar S. Regulation of E-cadherin expression and beta-catenin/Tcf transcriptional activity by the integrin-linked kinase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2004; 1691:1-15. [PMID: 15053919 DOI: 10.1016/j.bbamcr.2003.12.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2003] [Revised: 11/28/2003] [Accepted: 12/04/2003] [Indexed: 01/03/2023]
Abstract
Integrin-linked kinase (ILK) is a serine/threonine protein kinase which interacts with the cytoplasmic domains of beta1 and beta3 integrins. ILK structure and its localization at the focal adhesion allows it not only to interact with different structural proteins, but also to mediate many different signalling pathways. Extracellular matrices (ECM) and growth factors each stimulate ILK signalling. Constitutive activation of ILK in epithelial cells results in oncogenic phenotypes such as disruption of cell extracellular matrix and cell to cell interactions, suppression of suspension-induced apoptosis, and induction of anchorage independent cell growth and cell cycle progression. More specifically, pathological overexpression of ILK results in down-regulation of E-cadherin expression, and nuclear accumulation of beta-catenin, leading to the subsequent activation of the beta-catenin/Tcf transcription complex, the downstream components of the Wnt signalling pathway. Here we review the data implicating ILK in the regulation of these two signalling pathways, and discuss recent novel insights into the molecular basis and requirement of ILK in the process of epithelial to mesenchymal transformation (EMT).
Collapse
Affiliation(s)
- Arusha Oloumi
- British Columbia Cancer Agency and Jack Bell Research Centre, University of British Columbia, Vancouver Hospital, 2660 Oak St. Vancouver, BC, Canada V6H 3Z6
| | | | | |
Collapse
|
89
|
Bani MR, Nicoletti MI, Alkharouf NW, Ghilardi C, Petersen D, Erba E, Sausville EA, Liu ET, Giavazzi R. Gene expression correlating with response to paclitaxel in ovarian carcinoma xenografts. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.111.3.2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
We have investigated gene expression profiles of human ovarian carcinomas in vivo during Taxol® (paclitaxel) treatment and observed a difference in expression. Nude mice bearing 1A9 or 1A9PTX22 xenografts were given 60 mg/kg of paclitaxel. Therapeutic efficacy was achieved for 1A9, while 1A9PTX22 did not respond. Tumor tissues harvested 4 and 24 h after treatment were evaluated by cDNA microarray against untreated tumors. Paclitaxel caused the modulation of more genes in 1A9 than in 1A9PTX22 tumors, in accordance to their therapeutic response. Most gene expression alterations were detected 24 h after paclitaxel administration and affected genes involved in various biological functions including cell cycle regulation and cell proliferation (CDC2, CDKN1A, PLAB, and TOP2A), apoptosis (BNIP3 and PIG8), signal transduction and transcriptional regulation (ARF1, ATF2, FOS, GNA11, HDAC3, MADH2, SLUG, and SPRY4), fatty acid biosynthesis and sterol metabolism (FDPS, IDI1, LIPA, and SC5D), and IFN-mediated signaling (G1P3, IFI16, IFI27, IFITM1, and ISG15). The modulation of two representative genes, CDKN1A and TOP2A, was validated by Northern analyses on a panel of seven ovarian carcinoma xenograft models undergoing treatment with paclitaxel. We found that the changes in expression level of these genes was strictly associated with the responsiveness to paclitaxel. Our study shows the feasibility of obtaining gene expression profiles of xenografted tumor models as a result of drug exposure. This in turn might provide insights related to the drugs' action in vivo that will anticipate the response to treatment manifested by tumors and could be the basis for novel approaches to molecular pharmacodynamics.
Collapse
Affiliation(s)
- Maria Rosa Bani
- 1Mario Negri Institute for Pharmacological Research, Bergamo and Milan, Italy
| | | | - Nawal W. Alkharouf
- 2Advanced Technology Center, National Cancer Institute, Gaithersburg, MD
| | - Carmen Ghilardi
- 1Mario Negri Institute for Pharmacological Research, Bergamo and Milan, Italy
| | - David Petersen
- 2Advanced Technology Center, National Cancer Institute, Gaithersburg, MD
| | - Eugenio Erba
- 1Mario Negri Institute for Pharmacological Research, Bergamo and Milan, Italy
| | - Edward A. Sausville
- 3Developmental Therapeutics Program, National Cancer Institute, Rockville, MD; and
| | - Edison T. Liu
- 4Genome Institute of Singapore, National University of Singapore, Singapore
| | - Raffaella Giavazzi
- 1Mario Negri Institute for Pharmacological Research, Bergamo and Milan, Italy
| |
Collapse
|
90
|
Peinado H, Ballestar E, Esteller M, Cano A. Snail mediates E-cadherin repression by the recruitment of the Sin3A/histone deacetylase 1 (HDAC1)/HDAC2 complex. Mol Cell Biol 2004; 24:306-19. [PMID: 14673164 PMCID: PMC303344 DOI: 10.1128/mcb.24.1.306-319.2004] [Citation(s) in RCA: 574] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The transcription factor Snail has been described as a direct repressor of E-cadherin expression during development and carcinogenesis; however, the specific mechanisms involved in this process remain largely unknown. Here we show that mammalian Snail requires histone deacetylase (HDAC) activity to repress E-cadherin promoter and that treatment with trichostatin A (TSA) is sufficient to block the repressor effect of Snail. Moreover, overexpression of Snail is correlated with deacetylation of histones H3 and H4 at the E-cadherin promoter, and TSA treatment in Snail-expressing cells reverses the acetylation status of histones. Additionally, we demonstrate that Snail interacts in vivo with the E-cadherin promoter and recruits HDAC activity. Most importantly, we demonstrate an interaction between Snail, histone deacetylase 1 (HDAC1) and HDAC2, and the corepressor mSin3A. This interaction is dependent on the SNAG domain of Snail, indicating that the Snail transcription factor mediates the repression by recruitment of chromatin-modifying activities, forming a multimolecular complex to repress E-cadherin expression. Our results establish a direct causal relationship between Snail-dependent repression of E-cadherin and the modification of chromatin at its promoter.
Collapse
Affiliation(s)
- Hector Peinado
- Departamento de Bioquimica, Instituto de Investigaciones Biomédicas "Alberto Sols," Madrid, Spain
| | | | | | | |
Collapse
|
91
|
Abstract
The presence of a functional E-cadherin/catenin cell-cell adhesion complex is a prerequisite for normal development and maintenance of epithelial structures in the mammalian body. This implies that the acquisition of molecular abnormalities that disturb the expression or function of this complex is related to the development and progression of most, if not all, epithelial cell-derived tumors, i.e. carcinomas. E-cadherin downregulation is indeed correlated with malignancy parameters such as tumor progression, loss of differentiation, invasion and metastasis, and hence poor prognosis. Moreover, E-cadherin has been shown to be a potent invasion suppressor as well as a tumor suppressor. Disturbed expression profiles of the E-cadherin/catenin complex have been demonstrated in histological sections of many human tumor types. In different kinds of carcinomas, biallelic downregulation of the E-cadherin gene, resulting in tumor-restricted decrease or even complete loss of E-cadherin expression, appears to be caused by a variety of inactivation mechanisms. Gene deletion due to loss of heterozygosity of the CDH1 locus on 16q22.1 frequently occurs in many carcinoma types. However, somatic inactivating mutations resulting in aberrant E-cadherin expression by loss of both wild-type alleles is rare and restricted to only a few cancer types. A majority of carcinomas thus seems to show deregulated E-cadherin expression by other mechanisms. The present evidence proposes transcriptional repression as a powerful and recurrent molecular mechanism for silencing E-cadherin expression. The predominant mechanisms emerging in most carcinomas are hypermethylation of the E-cadherin promoter and expression of transrepressor molecules such as SIP1, Snail, and Slug that bind sequence elements in the proximal E-cadherin promoter. Interestingly, complex differential expression of other cadherins seems to be associated with loss of E-cadherin and to reinforce effects of this loss on tumor progression. Multiple agents can upregulate and stabilize the E-cadherin/catenin complex. Especially for those tumors with transcriptional and thus reversible downregulation of E-cadherin expression, these drug agents offer important therapeutic opportunities.
Collapse
|
92
|
McGhee L, Bryan J, Elliott L, Grimes HL, Kazanjian A, Davis JN, Meyers S. Gfi-1 attaches to the nuclear matrix, associates with ETO (MTG8) and histone deacetylase proteins, and represses transcription using a TSA-sensitive mechanism. J Cell Biochem 2003; 89:1005-18. [PMID: 12874834 DOI: 10.1002/jcb.10548] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gfi-1 and Gfi-1B can repress transcription and play important roles in hematopoietic cell survival and differentiation. Although these proteins are known to bind DNA through a C-terminal zinc-finger domain and may require an N-terminal SNAG domain (SNAIL/Gfi-1) to repress transcription, the mechanism by which Gfi-1 and Gfi-1B act is unknown. A first step towards understanding the mechanism by which these proteins repress transcription is to identify interacting proteins that could contribute to transcriptional repression. ETO (also termed MTG8), was first identified through its involvement in the (8;21) translocation associated with acute myelogenous leukemia. It attaches to the nuclear matrix and associates with histone deacetylases and the co-repressors N-CoR, SMRT, and mSin3A, and may act as a co-repressor for site-specific transcriptions factors. In this report we demonstrate that Gfi-1 interacts with ETO and related proteins both in vitro and in vivo and with histone deacetylase proteins in vivo. We observed that a portion of Gfi-1 and Gfi-1B associated with the nuclear matrix, as is the case with ETO. Moreover, Gfi-1 and ETO co-localize to punctate subnuclear structures. When co-expressed in mammalian cells, Gfi-1 associates with histone deacetylse-1 (HDAC-1), HDAC-2, and HDAC-3. These data identify ETO as a partner for Gfi-1 and Gfi-1B, and suggest that Gfi-1 proteins repress transcription through recruitment of histone deacetylase-containing complexes.
Collapse
Affiliation(s)
- Laura McGhee
- The Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-Shreveport, 1501 Kings Highway, Shreveport, LA 71130, USA
| | | | | | | | | | | | | |
Collapse
|
93
|
Domínguez D, Montserrat-Sentís B, Virgós-Soler A, Guaita S, Grueso J, Porta M, Puig I, Baulida J, Francí C, García de Herreros A. Phosphorylation regulates the subcellular location and activity of the snail transcriptional repressor. Mol Cell Biol 2003; 23:5078-89. [PMID: 12832491 PMCID: PMC162233 DOI: 10.1128/mcb.23.14.5078-5089.2003] [Citation(s) in RCA: 192] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2002] [Revised: 02/03/2003] [Accepted: 05/01/2003] [Indexed: 11/20/2022] Open
Abstract
The Snail gene product is a transcriptional repressor of E-cadherin expression and an inducer of the epithelial-to-mesenchymal transition in several epithelial tumor cell lines. This report presents data indicating that Snail function is controlled by its intracellular location. The cytosolic distribution of Snail depended on export from the nucleus by a CRM1-dependent mechanism, and a nuclear export sequence (NES) was located in the regulatory domain of this protein. Export of Snail was controlled by phosphorylation of a Ser-rich sequence adjacent to this NES. Modification of this sequence released the restriction created by the zinc finger domain and allowed nuclear export of the protein. The phosphorylation and subcellular distribution of Snail are controlled by cell attachment to the extracellular matrix. Suspended cells presented higher levels of phosphorylated Snail and an augmented extranuclear localization with respect to cells attached to the plate. These findings show the existence in tumor cells of an effective and fine-tuning nontranscriptional mechanism of regulation of Snail activity dependent on the extracellular environment.
Collapse
Affiliation(s)
- David Domínguez
- Unitat de Biologia Cel.lular i Molecular, Institut Municipal d'Investigació Mèdica, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Locascio A, Vega S, de Frutos CA, Manzanares M, Nieto MA. Biological potential of a functional human SNAIL retrogene. J Biol Chem 2002; 277:38803-9. [PMID: 12151403 DOI: 10.1074/jbc.m205358200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Snail genes encode zinc finger transcription factors required for the development of vertebrate and invertebrate embryos. They trigger epithelial to mesenchymal transitions (EMTs), thereby allowing epithelial cells to emigrate from their place of origin and form tissues such as the mesoderm and the neural crest. Snail genes are also involved in the EMTs responsible for the acquisition of invasiveness during tumor progression. This aspect of their activity is associated with their ability to directly repress E-cadherin transcription. Here we describe the existence of an active human Snail retrogene, inserted within an intron of a novel evolutionarily conserved gene and expressed in different human tissues and cell lines. Functional analyses in cell culture show that this retrogene maintains the potential to induce EMTs, conferring migratory and invasive properties to epithelial cells. In light of this data, we have renamed it SNAIL-like, a new player that must be considered in both physiological and pathological studies of SNAIL function in humans.
Collapse
Affiliation(s)
- Annamaria Locascio
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Av. Doctor Arce 37, 28002 Madrid, Spain
| | | | | | | | | |
Collapse
|
95
|
Abstract
The stem cell factor c-kit signaling pathway (SCF/c-kit) has been previously implicated in normal hematopoiesis, melanogenesis, and gametogenesis through the formation and migration of c-kit+ cells. These biologic functions are also determinants in epithelial–mesenchymal transitions during embryonic development governed by the Snail family of transcription factors. Here we show that the activation of c-kit by SCF specifically induces the expression of Slug, a Snail family member. Slug mutant mice have a cell-intrinsic defect with pigment deficiency, gonadal defect, and impairment of hematopoiesis. Kit+ cells derived from Slug mutant mice exhibit migratory defects similar to those of c-kit+ cells derived from SCF and c-kit mutant mice. Endogenous Slug is expressed in migratory c-kit+ cells purified from control mice but is not present in c-kit+cells derived from SCF mutant mice or in bone marrow cells from W/Wv mice, though Slug is present in spleen c-kit+ cells of W/Wv (mutants expressing c-kit with reduced surface expression and activity). SCF-induced migration was affected in primary c-kit+ cells purified from Slug−/− mice, providing evidence for a role of Slug in the acquisition of c-kit+ cells with ability to migrate. Slug may thus be considered a molecular target that contributes to the biologic specificity to the SCF/c-kit signaling pathway, opening up new avenues for stem cell mobilization.
Collapse
|
96
|
Abstract
The Snail superfamily of zinc-finger transcription factors is involved in processes that imply pronounced cell movements, both during embryonic development and in the acquisition of invasive and migratory properties during tumour progression. Different family members have also been implicated in the signalling cascade that confers left right identity, as well as in the formation of appendages, neural differentiation, cell division and cell survival.
Collapse
|
97
|
Carver EA, Jiang R, Lan Y, Oram KF, Gridley T. The mouse snail gene encodes a key regulator of the epithelial-mesenchymal transition. Mol Cell Biol 2001; 21:8184-8. [PMID: 11689706 PMCID: PMC99982 DOI: 10.1128/mcb.21.23.8184-8188.2001] [Citation(s) in RCA: 488] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Snail family genes encode DNA binding zinc finger proteins that act as transcriptional repressors. Mouse embryos deficient for the Snail (Sna) gene exhibit defects in the formation of the mesoderm germ layer. In Sna(-/-) mutant embryos, a mesoderm layer forms and mesodermal marker genes are induced but the mutant mesoderm is morphologically abnormal. Lacunae form within the mesoderm layer of the mutant embryos, and cells lining these lacunae retain epithelial characteristics. These cells resemble a columnar epithelium and have apical-basal polarity, with microvilli along the apical surface and intercellular electron-dense adhesive junctions that resemble adherens junctions. E-cadherin expression is retained in the mesoderm of the Sna(-/-) embryos. These defects are strikingly similar to the gastrulation defects observed in snail-deficient Drosophila embryos, suggesting that the mechanism of repression of E-cadherin transcription by Snail family proteins may have been present in the metazoan ancestor of the arthropod and mammalian lineages.
Collapse
Affiliation(s)
- E A Carver
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | | | | | |
Collapse
|
98
|
Savagner P. Leaving the neighborhood: molecular mechanisms involved during epithelial-mesenchymal transition. Bioessays 2001; 23:912-23. [PMID: 11598958 DOI: 10.1002/bies.1132] [Citation(s) in RCA: 541] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Several molecular mechanisms contribute directly and mechanically to the loss of epithelial phenotype. During epithelial-mesenchymal transition (EMT), adherens junctions and desmosomes are at least partially dissociated. At the same time, a massive cytoskeleton reorganization takes place, involving the rho family and the remodeling of the actin microfilament mesh. Numerous pathways have been described in vitro that control phenotype transition in specific cell models. In vivo developmental studies suggest that transcriptional control, activated by a specific pathway involving Ras, Src and potentially the Wnt pathway, is an essential step. Recent functional and localization experiments indicate that the slug/snail family of transcription factors functions overall as an epithelial phenotype repressor and could represent a key EMT contributor.
Collapse
Affiliation(s)
- P Savagner
- Equipe Genome et Cancer, UMR CNRS 5535, CRLC Val d'Aurelle-Paul Larmarque, 34298 Montpellier, cedex 5, France.
| |
Collapse
|
99
|
Abstract
E2A-HLF, the chimeric fusion protein resulting from the leukemogenic translocation t(17;19), appears to employ evolutionarily conserved signaling cascades for its transforming and antiapoptotic functions. These arise from both impairment of normal E2A function and activation of a survival pathway triggered through the HLF bZip DNA binding and dimerization domain. Recent reports identify wild-type E2A as a tumor suppressor in T lymphocytes. Moreover, E2A-HLF has been shown to activate SLUG, a mammalian homologue of the cell death specification protein CES-1 in Caenorhabditis elegans, which appears to regulate an evolutionarily conserved cell survival program. Recently, several key mouse models have been generated, enabling further elucidation of these pathways on a molecular genetic level in vivo. In this review, we discuss the characteristics of both components of the fusion protein with regard to their contribution to the regulation of cell fate and the oncogenic potential of E2A-HLF.
Collapse
Affiliation(s)
- M G Seidel
- Pediatric Oncology Department, Dana-Farber Cancer Institute, 44 Binney Street, M-630, Boston, Massachusetts, MA 02115, USA
| | | |
Collapse
|
100
|
Yin X, Landay MF, Han W, Levitan ES, Watkins SC, Levenson RM, Farkas DL, Prochownik EV. Dynamic in vivo interactions among Myc network members. Oncogene 2001; 20:4650-64. [PMID: 11498788 DOI: 10.1038/sj.onc.1204606] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2001] [Revised: 05/03/2001] [Accepted: 05/09/2001] [Indexed: 11/09/2022]
Abstract
Members of the Myc oncoprotein network (c-Myc, Max, and Mad) play important roles in proliferation, differentiation, and apoptosis. We expressed chimeric green fluorescent protein (GFP) fusions of c-Myc, Max, and three Mad proteins in fibroblasts. Individually, c-Myc and Mad proteins localized in subnuclear speckles, whereas Max assumed a homogeneous nuclear pattern. These distributions were co-dominant and dynamic, however, as each protein assumed the pattern of its heterodimeric partner when the latter was co-expressed at a higher level. Deletion mapping of two Mad members, Mad1 and Mxi1, demonstrated that the domains responsible for nuclear localization and speckling are separable. A non-speckling Mxi1 mutant was also less effective as a transcriptional repressor than wild-type Mxi1. c-Myc nuclear speckles were distinct from SC-35 domains involved in mRNA processing. However, in the presence of co-expressed Max, c-Myc, but not Mad, co-localized to a subset of SC-35 loci. These results show that Myc network proteins comprise dynamic subnuclear structures and behave co-dominantly when co-expressed with their normal heterodimerization partners. In addition, c-Myc-Max heterodimers, but not Max-Mad heterodimers, localize to foci actively engaged in pre-mRNA transcription/processing. These findings suggest novel means by which Myc network members promote transcriptional activation or repression.
Collapse
Affiliation(s)
- X Yin
- Section of Hematology/Oncology, Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, PA 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|