301
|
Lin CY, Tsai PH, Kandaswami CC, Lee PP, Huang CJ, Hwang JJ, Lee MT. Matrix metalloproteinase-9 cooperates with transcription factor Snail to induce epithelial-mesenchymal transition. Cancer Sci 2011; 102:815-27. [PMID: 21219539 DOI: 10.1111/j.1349-7006.2011.01861.x] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Chun-Yu Lin
- Institute of Biochemical Sciences, School of Life Sciences, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
302
|
Abstract
The transcriptional inhibitor Snail is a critical regulator for epithelial-mesenchymal transition (EMT). Although low oxygen induces Snail transcription, thereby stimulating EMT, a direct role of hypoxia-inducible factor (HIF) in this process remains to be demonstrated. Here we show that hypoxia induces the expression of Snail via HIF. In silico analysis identified a potential hypoxia-response element (HRE) close to the minimal promoter of the human and mouse genome of the snail gene. Gel shift assays demonstrated that a specific hypoxia-inducible complex is formed with the putative HRE and that the complex contains HIF proteins. ChIP assays confirmed the interaction of HIF proteins with the putative HRE in vivo. Reporter gene analyses showed that the putative HRE responds to hypoxia in its natural position as well as in front of a heterologous promoter and that the HRE is directly activated by HIF-1α or HIF-2α. HIF knockdown with siRNA at 2% oxygen and overexpression of an oxygen-insensitive HIF (HIF-ΔODD) mutant at 21% oxygen showed that HIF regulates Snail activation and subsequent cell migration. Our findings identify snail as a HIF target gene and provide novel insights into the regulation of snail and hypoxia-induced EMT.
Collapse
Affiliation(s)
- Daochun Luo
- Physiology and Experimental Medicine, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8 Canada
| | | | | | | |
Collapse
|
303
|
Baritaki S, Huerta-Yepez S, Sahakyan A, Karagiannides I, Bakirtzi K, Jazirehi A, Bonavida B. Mechanisms of nitric oxide-mediated inhibition of EMT in cancer: inhibition of the metastasis-inducer Snail and induction of the metastasis-suppressor RKIP. Cell Cycle 2010; 9:4931-40. [PMID: 21150329 DOI: 10.4161/cc.9.24.14229] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The role of nitric oxide (NO) in cancer has been controversial and is based on the levels of NO and the responsiveness of the tumor type. It remains unclear whether NO can inhibit the epithelial to mesenchymal transition (EMT) in cancer cells. EMT induction is mediated, in part, by the constitutive activation of the metastasis-inducer transcription factor, Snail and EMT can be inhibited by the metastasis-suppressor Raf-1 kinase inhibitor protein (RKIP) and E-cadherin. Snail is transcriptionally regulated by NF-κB and in turn, Snail represses RKIP transcription. Hence, we hypothesized that high levels of NO, that inhibit NF-κB activity, may also inhibit Snail and induce RKIP and leading to inhibition of EMT. We show that treatment of human prostate metastatic cell lines with the NO donor, DETANONOate, inhibits EMT and reverses both the mesenchymal phenotype and the cell invasive properties. Further, treatment with DETANONOate inhibits Snail expression and DNA-binding activity in parallel with the upregulation of RKIP and E-cadherin protein levels. The pivotal roles of Snail inhibition and RKIP induction in DETANONOate-mediated inhibition of EMT were corroborated by both Snail silencing by siRNA and by ectopic expression of RKIP. The in vitro findings were validated in vivo in mice bearing PC-3 xenografts and treated with DETANONOate. The present findings show, for the first time, the novel role of high subtoxic concentrations of NO in the inhibition of EMT. Thus, NO donors may exert therapeutic activities in the reversal of EMT and metastasis.
Collapse
Affiliation(s)
- Stavroula Baritaki
- Microbiology, Immunology and Molecular Genetics, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | | | | | | | | | | | | |
Collapse
|
304
|
Qu Y, Wang J, Ray PS, Guo H, Huang J, Shin-Sim M, Bukoye BA, Liu B, Lee AV, Lin X, Huang P, Martens JW, Giuliano AE, Zhang N, Cheng NH, Cui X. Thioredoxin-like 2 regulates human cancer cell growth and metastasis via redox homeostasis and NF-κB signaling. J Clin Invest 2010; 121:212-25. [PMID: 21123948 DOI: 10.1172/jci43144] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 09/29/2010] [Indexed: 12/14/2022] Open
Abstract
Cancer cells have an efficient antioxidant system to counteract their increased generation of ROS. However, whether this ability to survive high levels of ROS has an important role in the growth and metastasis of tumors is not well understood. Here, we demonstrate that the redox protein thioredoxin-like 2 (TXNL2) regulates the growth and metastasis of human breast cancer cells through a redox signaling mechanism. TXNL2 was found to be overexpressed in human cancers, including breast cancers. Knockdown of TXNL2 in human breast cancer cell lines increased ROS levels and reduced NF-κB activity, resulting in inhibition of in vitro proliferation, survival, and invasion. In addition, TXNL2 knockdown inhibited tumorigenesis and metastasis of these cells upon transplantation into immunodeficient mice. Furthermore, analysis of primary breast cancer samples demonstrated that enhanced TXNL2 expression correlated with metastasis to the lung and brain and with decreased overall patient survival. Our studies provided insight into redox-based mechanisms underlying tumor growth and metastasis and suggest that TXNL2 could be a target for treatment of breast cancer.
Collapse
Affiliation(s)
- Ying Qu
- Department of Surgery, Ruijin Hospital, Institute of Digestive Surgery, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
305
|
Brandl M, Seidler B, Haller F, Adamski J, Schmid RM, Saur D, Schneider G. IKK(α) controls canonical TGF(ß)-SMAD signaling to regulate genes expressing SNAIL and SLUG during EMT in panc1 cells. J Cell Sci 2010; 123:4231-9. [PMID: 21081648 DOI: 10.1242/jcs.071100] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a crucial step in tumor progression, and the TGFβ-SMAD signaling pathway is an inductor of EMT in many tumor types. One hallmark of EMT is downregulation of the adherens junction protein E-cadherin, a process mediated by transcription factors such as the zinc fingers SNAIL and SLUG. Here, we report that the catalytic IκB kinase (IKK) subunit IKKα is necessary for the silencing of E-cadherin in a Panc1 cell model of TGFβ-SMAD-mediated EMT, independently of NFκB. IKKα regulates canonical TGFβ-SMAD signaling by interacting with SMAD3 and controlling SMAD complex formation on DNA. Furthermore, we demonstrate that the TGFβ-IKKα-SMAD signaling pathway induces transcription of the genes encoding SNAIL and SLUG. In addition, we demonstrate that IKKα also modulates canonical TGFβ-SMAD signaling in human MDA-MB231 breast cancer cells, arguing for a more general impact of IKKα on the control of TGFβ-SMAD signaling. Taken together, these findings indicate that IKKα contributes to the tumor-promoting function of the TGFβ-SMAD signaling pathway in particular cancers.
Collapse
Affiliation(s)
- Martina Brandl
- II. Medizinische Klinik, Technische Universität München, Ismaninger Strasse 22, 81675 München, Germany
| | | | | | | | | | | | | |
Collapse
|
306
|
Shi MF, Jiao J, Lu WG, Ye F, Ma D, Dong QG, Xie X. Identification of cancer stem cell-like cells from human epithelial ovarian carcinoma cell line. Cell Mol Life Sci 2010; 67:3915-25. [PMID: 20549538 PMCID: PMC11115598 DOI: 10.1007/s00018-010-0420-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 04/23/2010] [Accepted: 05/25/2010] [Indexed: 12/21/2022]
Abstract
Cancer stem cells (CSCs) play an important role in the development, invasion, and drug resistance of carcinoma, but the exact phenotype and characteristics of ovarian CSCs are still disputable. In this study, we identified cancer stem cell-like cells (CSC-LCs) and investigated their characteristics from the ovarian adenocarcinoma cell line 3AO. Our results showed that CSC-LCs were enriched in sphere-forming test and highly expressed CD44(+)CD24⁻. The spheres and CD24⁻ cells possessed strong tumorigenic ability by transplantation into nonobese diabetic/severe combined immunodeficient mice. CD44(+)CD24⁻ cells expressed stem cell markers and differentiated to CD44(+)CD24(+) cells by immunofluorescence assay and fluorescence-activated cell-sorting analysis. In vitro experiments verified that CD44(+)CD24⁻ cells were markedly resistant to carboplatin and paclitaxol. In conclusion, our study identifies the CD44(+)CD24⁻ phenotype, self-renewal, high tumorigenicity, differentiation potential, and drug resistance of ovarian CSC-LCs. Our findings may provide the evidence needed to explore a new strategy in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- M. F. Shi
- Department of Gynecologic Oncology, Women’s Hospital, School of Medicine, Women’s Reproductive Health Laboratory of Zhejiang Province, Zhejiang University, 2 Xueshi Road, Hangzhou, Zhejiang China
| | - J. Jiao
- Department of Gynecologic Oncology, Women’s Hospital, School of Medicine, Women’s Reproductive Health Laboratory of Zhejiang Province, Zhejiang University, 2 Xueshi Road, Hangzhou, Zhejiang China
| | - W. G. Lu
- Department of Gynecologic Oncology, Women’s Hospital, School of Medicine, Women’s Reproductive Health Laboratory of Zhejiang Province, Zhejiang University, 2 Xueshi Road, Hangzhou, Zhejiang China
| | - F. Ye
- Department of Gynecologic Oncology, Women’s Hospital, School of Medicine, Women’s Reproductive Health Laboratory of Zhejiang Province, Zhejiang University, 2 Xueshi Road, Hangzhou, Zhejiang China
| | - D. Ma
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Q. G. Dong
- Shanghai Cancer Institute, Shanghai Jiao Tong University, 2200 Xietu Road, Shanghai, China
| | - X. Xie
- Department of Gynecologic Oncology, Women’s Hospital, School of Medicine, Women’s Reproductive Health Laboratory of Zhejiang Province, Zhejiang University, 2 Xueshi Road, Hangzhou, Zhejiang China
| |
Collapse
|
307
|
Gil D, Ciołczyk-Wierzbicka D, Dulińska-Litewka J, Zwawa K, McCubrey JA, Laidler P. The mechanism of contribution of integrin linked kinase (ILK) to epithelial-mesenchymal transition (EMT). ACTA ACUST UNITED AC 2010; 51:195-207. [PMID: 21035499 DOI: 10.1016/j.advenzreg.2010.09.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 09/05/2010] [Indexed: 12/22/2022]
Abstract
Integrin linked kinase (ILK) is ubiquitously expressed serine/threonine protein kinase, a binding partner of β1 and β3 integrin subunit as a cytoplasmic effector of integrin receptors that functionally links them to the actin cytoskeleton.We postulate that ILK is important enzyme involved in epithelial-mesenchymal transition (EMT) a critical event in the process of cancer progression. Commonly used EMT molecular markers include among others increased expression of N-cadherin and vimentin, nuclear localization of β-catenin, and the decrease of E-cadherin synthesis. In this study we were able to show that N-cadherin expression in melanoma cells is dependent on ILK signaling and the translocation of β-catenin to the nucleus. Silencing of ILK expression by siRNA significantly inhibited the stabilization and subsequent nuclear translocation of β-catenin and the expression of N-cadherin, a crucial molecule in the EMT, which facilitates association with fibroblast and endothelial cells during invasion of various cancers. The results allow to cautiously speculate on the important role of ILK in the cross-talk between integrins and cadherins accompanying EMT in melanoma.
Collapse
Affiliation(s)
- Dorota Gil
- Chair of Medical Biochemistry Jagiellonian University Medical College, Kraków, Poland.
| | | | | | | | | | | |
Collapse
|
308
|
Bonavida B, Baritaki S. Dual role of NO donors in the reversal of tumor cell resistance and EMT: Downregulation of the NF-κB/Snail/YY1/RKIP circuitry. Nitric Oxide 2010; 24:1-7. [PMID: 20933602 DOI: 10.1016/j.niox.2010.10.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 09/29/2010] [Accepted: 10/01/2010] [Indexed: 12/16/2022]
Abstract
Several studies have implicated the role of Nitric Oxide (NO) in the regulation of tumor cell behavior and have shown that NO either promotes or inhibits tumorigenesis. These conflicting findings have been resolved, in part, by the levels of NO used such that low levels promote tumor growth and high levels inhibit tumor growth. Our studies have focused on the use of high levels of NO provided primarily by the NO donor, DETANONOate. We have shown that treatment of resistant tumor cells with DETANONOate sensitizes them to apoptosis by both chemotherapeutic drugs and cytotoxic immunotherapeutic ligands. The underlying mechanisms by which NO sensitizes tumor cells to apoptosis were shown to be regulated, in part, by NO-mediated inhibition of the NF-κB survival/anti-apoptotic pathways and downstream of NF-κB by inhibition of the transcription factor Yin Yang 1 (YY1). In addition to NO-induced sensitization to apoptosis, we have also shown that NO induced the expression of the metastasis-suppressor/immunosurveillance cancer gene product, Raf-1 kinase inhibitor protein (RKIP). Overexpression of RKIP mimics NO in tumor cells-induced sensitization to apoptosis. The induction of RKIP by NO was the result of the inhibition of the RKIP repressor, Snail, downstream of NF-κB. These findings established the presence of a dysregulated NF-κB/Snail/YY1/ RKIP circuitry in resistance and that treatment with NO modifies this loop in tumor cells in favor of the inhibition of tumor cell survival and the response to cytotoxic drugs. Noteworthy, the NF-κB/Snail/YY1/RKIP loop consists of gene products that regulate the epithelial to mesenchymal transition (EMT) and, thus, tumor metastasis. Hence, we have found that treatment of metastatic cancer cell lines with DETANONOate inhibited the EMT phenotype, through both the inhibition of the metastasis-inducers, NF-κB and Snail and the induction of the metastasis-suppressor, RKIP. Altogether, the above findings establish, for the first time, the dual role of high levels of NO in the sensitization of tumor cells to apoptotic stimuli as well as inhibition of EMT. Hence, NO donors may be considered as novel potential therapeutic agents with dual roles in the treatment of patients with refractory cancer and in the prevention of the initiation of the metastatic cascade via EMT.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, Johnson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA.
| | | |
Collapse
|
309
|
Wang Z, Li Y, Kong D, Sarkar FH. The role of Notch signaling pathway in epithelial-mesenchymal transition (EMT) during development and tumor aggressiveness. Curr Drug Targets 2010; 11:745-51. [PMID: 20041844 DOI: 10.2174/138945010791170860] [Citation(s) in RCA: 254] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 12/17/2009] [Indexed: 02/07/2023]
Abstract
The Notch signaling pathway maintains a balance between cell proliferation and apoptosis, and thus it is believed that Notch signaling pathways may play an important role in the development and progression of several malignancies. However, the functions of Notch signaling in EMT are largely unknown. This mini review describes the role of Notch signaling pathway in EMT, and cataloging how its deregulation is involved in EMT and tumor aggressiveness. Further attempts have been made to summarize the role of several chemopreventive agents that could be useful for targeted inactivation of Notch signaling, and thus it may cause reversal of EMT, which could become a novel approach for cancer prevention and treatment.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | | | | | | |
Collapse
|
310
|
Wang Z, Li Y, Sarkar FH. Signaling mechanism(s) of reactive oxygen species in Epithelial-Mesenchymal Transition reminiscent of cancer stem cells in tumor progression. Curr Stem Cell Res Ther 2010; 5:74-80. [PMID: 19951255 DOI: 10.2174/157488810790442813] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 10/16/2009] [Indexed: 12/22/2022]
Abstract
Reactive oxygen species (ROS) are known to serve as a second messenger in the intracellular signal transduction pathway for a variety of cellular processes, including inflammation, cell cycle progression, apoptosis, aging and cancer. Recently, ROS have been found to be associated with tumor metastasis involving the processes of tumor cell migration, invasion and angiogenesis. Emerging evidence also suggests that Epithelial-Mesenchymal Transition (EMT), a process that is reminiscent of cancer stem cells, is an important step toward tumor invasion and metastasis, and intimately involved in de novo and acquired drug resistance. In light of recent advances, we are summarizing the role of ROS in EMT by cataloging how its deregulation is involved in EMT and tumor aggressiveness. Further attempts have been made to summarize the role of several chemopreventive agents that could be useful for targeted inactivation of ROS, suggesting that many natural agents could be useful for the reversal of EMT, which would become a novel approach for the prevention of tumor progression and/or treatment of human malignancies especially by killing EMT-type cells that shares similar characteristics with cancer stem cells.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
311
|
Sume SS, Kantarci A, Lee A, Hasturk H, Trackman PC. Epithelial to mesenchymal transition in gingival overgrowth. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:208-18. [PMID: 20489142 PMCID: PMC2893664 DOI: 10.2353/ajpath.2010.090952] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/24/2010] [Indexed: 12/23/2022]
Abstract
Epithelial to mesenchymal transition (EMT) occurs normally in development. In pathology, EMT drives cancer and fibrosis. Medication with phenytoin, nifedipine, and cyclosporine-A often causes gingival overgrowth. Based partly on the histopathology of gingival overgrowth, the present study investigates the hypothesis that EMT could contribute to its development. We found that phenytoin-induced human gingival overgrowth tissues, the most fibrotic drug-induced variety, contain diminished epithelial E-cadherin expression, whereas fibroblast-specific protein-1 (FSP-1) and alphavbeta6 integrin levels are up-regulated. In connective tissue stroma, fibronectin and alternatively spliced fibronectin extra type III domain A (FN-ED-A) levels are increased in overgrowth lesions. Transforming growth factor (TGF)-beta1 treatment of primary human gingival epithelial cells cultured in transwell plates resulted in inhibited barrier function as determined by reduced electrical resistance, paracellular permeability assays, and cell surface E-cadherin expression. Moreover, TGF-beta1 altered the expression of other markers of EMT determined at the mRNA and protein levels: E-cadherin decreased, whereas SLUG, fibronectin, matrix metalloproteinase (MMP)2, MMP9, and MMP13 increased. Nifedipine- and cyclosporine A-induced gingival overgrowth tissues similarly contain diminished E-cadherin and elevated levels of FSP-1 and fibronectin, but normal levels of alphavbeta6 integrin. In summary, data in vitro support that human gingival epithelial cells undergo functional and gene expression changes consistent with EMT in response to TGF-beta1, and in vivo studies show that important EMT markers occur in clinical gingival overgrowth tissues. These findings support the hypothesis that EMT likely occurs in drug-induced gingival overgrowth.
Collapse
Affiliation(s)
- Siddika Selva Sume
- Department of Periodontology and Oral Biology, Boston University, Henry M. Goldman School of Dental Medicine, 700 Albany Street W-201, Boston, MA 02118, USA
| | | | | | | | | |
Collapse
|
312
|
Epstein-Barr virus-encoded LMP2A induces an epithelial-mesenchymal transition and increases the number of side population stem-like cancer cells in nasopharyngeal carcinoma. PLoS Pathog 2010; 6:e1000940. [PMID: 20532215 PMCID: PMC2880580 DOI: 10.1371/journal.ppat.1000940] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 05/05/2010] [Indexed: 12/29/2022] Open
Abstract
It has been recently reported that a side population of cells in nasopharyngeal carcinoma (NPC) displayed characteristics of stem-like cancer cells. However, the molecular mechanisms underlying the modulation of such stem-like cell populations in NPC remain unclear. Epstein-Barr virus was the first identified human tumor virus to be associated with various malignancies, most notably NPC. LMP2A, the Epstein-Barr virus encoded latent protein, has been reported to play roles in oncogenic processes. We report by immunostaining in our current study that LMP2A is overexpressed in 57.6% of the nasopharyngeal carcinoma tumors sampled and is mainly localized at the tumor invasive front. We found also in NPC cells that the exogenous expression of LMP2A greatly increases their invasive/migratory ability, induces epithelial-mesenchymal transition (EMT)-like cellular marker alterations, and stimulates stem cell side populations and the expression of stem cell markers. In addition, LMP2A enhances the transforming ability of cancer cells in both colony formation and soft agar assays, as well as the self-renewal ability of stem-like cancer cells in a spherical culture assay. Additionally, LMP2A increases the number of cancer initiating cells in a xenograft tumor formation assay. More importantly, the endogenous expression of LMP2A positively correlates with the expression of ABCG2 in NPC samples. Finally, we demonstrate that Akt inhibitor (V) greatly decreases the size of the stem cell side populations in LMP2A-expressing cells. Taken together, our data indicate that LMP2A induces EMT and stem-like cell self-renewal in NPC, suggesting a novel mechanism by which Epstein-Barr virus induces the initiation, metastasis and recurrence of NPC.
Collapse
|
313
|
NF-κB signaling modulation by EBV and KSHV. Trends Microbiol 2010; 18:248-57. [DOI: 10.1016/j.tim.2010.04.001] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 03/24/2010] [Accepted: 04/02/2010] [Indexed: 12/12/2022]
|
314
|
Bonnomet A, Brysse A, Tachsidis A, Waltham M, Thompson EW, Polette M, Gilles C. Epithelial-to-mesenchymal transitions and circulating tumor cells. J Mammary Gland Biol Neoplasia 2010; 15:261-73. [PMID: 20449641 DOI: 10.1007/s10911-010-9174-0] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 04/20/2010] [Indexed: 12/13/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) phenomena endow epithelial cells with enhanced migratory and invasive potential, and as such, have been implicated in many physiological and pathological processes requiring cell migration/invasion. Although their involvement in the metastatic cascade is still a subject of debate, data are accumulating to demonstrate the existence of EMT phenotypes in primary human tumors, describe enhanced metastatic potential of EMT derivatives in animal models, and report EMT attributes in circulating tumor cells (CTCs). The relationships between EMT and CTCs remain largely unexplored, and we review here in vitro and in vivo data supporting a putative role of EMT processes in CTC generation and survival.
Collapse
Affiliation(s)
- Arnaud Bonnomet
- Laboratory of Tumor and Developmental Biology, Liège University, GIGA - Cancer, C.H.U. Sart-Tilman, Tour de Pathologie B23, 4000 Liège, Belgium
| | | | | | | | | | | | | |
Collapse
|
315
|
van de Sluis B, Mao X, Zhai Y, Groot AJ, Vermeulen JF, van der Wall E, van Diest PJ, Hofker MH, Wijmenga C, Klomp LW, Cho KR, Fearon ER, Vooijs M, Burstein E. COMMD1 disrupts HIF-1alpha/beta dimerization and inhibits human tumor cell invasion. J Clin Invest 2010; 120:2119-30. [PMID: 20458141 PMCID: PMC2877941 DOI: 10.1172/jci40583] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Accepted: 03/17/2010] [Indexed: 11/17/2022] Open
Abstract
The gene encoding COMM domain-containing 1 (COMMD1) is a prototypical member of the COMMD gene family that has been shown to inhibit both NF-kappaB- and HIF-mediated gene expression. NF-kappaB and HIF are transcription factors that have been shown to play a role in promoting tumor growth, survival, and invasion. In this study, we demonstrate that COMMD1 expression is frequently suppressed in human cancer and that decreased COMMD1 expression correlates with a more invasive tumor phenotype. We found that direct repression of COMMD1 in human cell lines led to increased tumor invasion in a chick xenograft model, while increased COMMD1 expression in mouse melanoma cells led to decreased lung metastasis in a mouse model. Decreased COMMD1 expression also correlated with increased expression of genes known to promote cancer cell invasiveness, including direct targets of HIF. Mechanistically, our studies show that COMMD1 inhibits HIF-mediated gene expression by binding directly to the amino terminus of HIF-1alpha, preventing its dimerization with HIF-1beta and subsequent DNA binding and transcriptional activation. Altogether, our findings demonstrate a role for COMMD1 in tumor invasion and provide a detailed mechanism of how this factor regulates the HIF pathway in cancer cells.
Collapse
Affiliation(s)
- Bart van de Sluis
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xicheng Mao
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yali Zhai
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Arjan J. Groot
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jeroen F. Vermeulen
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Elsken van der Wall
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Paul J. van Diest
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Marten H. Hofker
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Cisca Wijmenga
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Leo W. Klomp
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kathleen R. Cho
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Eric R. Fearon
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Marc Vooijs
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ezra Burstein
- Complex Genetics Section, Division of Biomedical Genetics, Department of Medical Genetics, and
Laboratory for Metabolic and Endocrine Diseases, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Pathology and Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Department of Pathology and
Department of Internal Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
Maastricht Radiation Oncology (MAASTRO Laboratory), GROW — School for Oncology and Developmental Biology, University Medical Centre Maastricht, Maastricht, Netherlands.
Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
316
|
Previdi S, Maroni P, Matteucci E, Broggini M, Bendinelli P, Desiderio MA. Interaction between human-breast cancer metastasis and bone microenvironment through activated hepatocyte growth factor/Met and β-catenin/Wnt pathways. Eur J Cancer 2010; 46:1679-91. [DOI: 10.1016/j.ejca.2010.02.036] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 01/21/2010] [Accepted: 02/23/2010] [Indexed: 11/15/2022]
|
317
|
Gavert N, Ben-Shmuel A, Lemmon V, Brabletz T, Ben-Ze'ev A. Nuclear factor-kappaB signaling and ezrin are essential for L1-mediated metastasis of colon cancer cells. J Cell Sci 2010; 123:2135-43. [PMID: 20501702 DOI: 10.1242/jcs.069542] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hyperactivation of beta-catenin-T-cell-factor (TCF)-regulated gene transcription is a hallmark of colorectal cancer (CRC). The cell-neural adhesion molecule L1CAM (hereafter referred to as L1) is a target of beta-catenin-TCF, exclusively expressed at the CRC invasive front in humans. L1 overexpression in CRC cells increases cell growth and motility, and promotes liver metastasis. Genes induced by L1 are also expressed in human CRC tissue but the mechanisms by which L1 confers metastasis are still unknown. We found that signaling by the nuclear factor kappaB (NF-kappaB) is essential, because inhibition of signaling by the inhibitor of kappaB super repressor (IkappaB-SR) blocked L1-mediated metastasis. Overexpression of the NF-kappaB p65 subunit was sufficient to increase CRC cell proliferation, motility and metastasis. Binding of the L1 cytodomain to ezrin - a cytoskeleton-crosslinking protein - is necessary for metastasis because when binding to L1 was interrupted or ezrin gene expression was suppressed with specific shRNA, metastasis did not occur. L1 and ezrin bound to and mediated the phosphorylation of IkappaB. We also observed a complex containing IkappaB, L1 and ezrin in the juxtamembrane region of CRC cells. Furthermore, we found that L1, ezrin and phosphorylated p65 are co-expressed at the invasive front in human CRC tissue, indicating that L1-mediated activation of NF-kappaB signaling involving ezrin is a major route of CRC progression.
Collapse
Affiliation(s)
- Nancy Gavert
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | |
Collapse
|
318
|
Bauerle KT, Schweppe RE, Haugen BR. Inhibition of nuclear factor-kappa B differentially affects thyroid cancer cell growth, apoptosis, and invasion. Mol Cancer 2010; 9:117. [PMID: 20492683 PMCID: PMC2887796 DOI: 10.1186/1476-4598-9-117] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Accepted: 05/21/2010] [Indexed: 01/28/2023] Open
Abstract
Background Nuclear factor-κB (NF-κB) is constitutively activated in many cancers and plays a key role in promoting cell proliferation, survival, and invasion. Our understanding of NF-κB signaling in thyroid cancer, however, is limited. In this study, we have investigated the role of NF-κB signaling in thyroid cancer cell proliferation, invasion, and apoptosis using selective genetic inhibition of NF-κB in advanced thyroid cancer cell lines. Results Three pharmacologic inhibitors of NF-κB differentially inhibited growth in a panel of advanced thyroid cancer cell lines, suggesting that these NF-κB inhibitors may have off-target effects. We therefore used a selective genetic approach to inhibit NF-κB signaling by overexpression of a dominant-negative IκBα (mIκBα). These studies revealed decreased cell growth in only one of five thyroid cancer cell lines (8505C), which occurred through a block in the S-G2/M transition. Resistance to TNFα-induced apoptosis was observed in all cell lines, likely through an NF-κB-dependent mechanism. Inhibition of NF-κB by mIκBα sensitized a subset of cell lines to TNFα-induced apoptosis. Sensitive cell lines displayed sustained activation of the stress-activated protein kinase/c-Jun NH2-terminal kinase (SAPK/JNK) pathway, defining a potential mechanism of response. Finally, NF-κB inhibition by mIκBα expression differentially reduced thyroid cancer cell invasion in these thyroid cancer cell lines. Sensitive cell lines demonstrated approximately a two-fold decrease in invasion, which was associated with differential expression of MMP-13. MMP-9 was reduced by mIκBα expression in all cell lines tested. Conclusions These data indicate that selective inhibition of NF-κB represents an attractive therapeutic target for the treatment of advanced thyroid. However, it is apparent that global regulation of thyroid cancer cell growth and invasion is not achieved by NF-κB signaling alone. Instead, our findings suggest that other important molecular processes play a critical role in defining the extent of NF-κB function within cancer cells.
Collapse
Affiliation(s)
- Kevin T Bauerle
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado, Research Complex I, South Tower, Mail Stop 8106, 12801 East 17th Avenue, PO Box 6511, Aurora, CO 80045, USA
| | | | | |
Collapse
|
319
|
Maier HJ, Schmidt-Strassburger U, Huber MA, Wiedemann EM, Beug H, Wirth T. NF-kappaB promotes epithelial-mesenchymal transition, migration and invasion of pancreatic carcinoma cells. Cancer Lett 2010; 295:214-28. [PMID: 20350779 DOI: 10.1016/j.canlet.2010.03.003] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 02/26/2010] [Accepted: 03/04/2010] [Indexed: 11/29/2022]
Abstract
The transcription factor NF-kappaB is constitutively active in pancreatic adenocarcinoma. Here we explore the contribution of NF-kappaB to the malignant phenotype of pancreatic cancer cells in addition to its anti-apoptotic role. Block of NF-kappaB signalling by non-destructible IkappaBalpha rendered cells resistant to TGF-beta-induced epithelial-mesenchymal transition (EMT). In contrast, NF-kappaB activation by TNF-alpha or expression of constitutively active IKK2 induced an EMT-phenotype with up-regulation of vimentin and ZEB1, and down-regulation of E-cadherin. EMT could also be induced in cells with defective TGF-beta signalling. Functional assays demonstrated reduced or strongly enhanced migration and invasion upon NF-kappaB inhibition or activation, respectively.
Collapse
Affiliation(s)
- Harald J Maier
- Institute of Physiological Chemistry, University of Ulm, Germany
| | | | | | | | | | | |
Collapse
|
320
|
Arachidonic acid promotes epithelial-to-mesenchymal-like transition in mammary epithelial cells MCF10A. Eur J Cell Biol 2010; 89:476-88. [PMID: 20207443 DOI: 10.1016/j.ejcb.2009.12.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 12/19/2009] [Accepted: 12/21/2009] [Indexed: 11/22/2022] Open
Abstract
Epidemiological studies and animal models suggest an association between high levels of dietary fat intake and an increased risk of breast cancer. Cancer progression requires the development of metastasis, which is characterized by an increase in cell motility and invasion. Epithelial-to-mesenchymal transition (EMT) is a process, by which epithelial cells are transdifferentiated to a more mesenchymal state. A similar process takes place during tumor progression, when carcinoma cells stably or transiently lose epithelial polarities and acquire a mesenchymal phenotype. Arachidonic acid (AA) is a fatty acid that mediates cellular processes, such as cell survival, angiogenesis, chemotaxis, mitogenesis, migration and apoptosis. However, the role of AA on the EMT process in human mammary epithelial cells remains to be studied. We demonstrate here that AA promotes an increase in vimentin and N-cadherin expression, MMP-9 secretion, a decrease in E-cadherin junctional levels, and the activation of FAK, Src and NF-kappaB in MCF10A cells. Furthermore, AA also promotes cell migration in an Src kinase activity-dependent fashion. In conclusion, our results demonstrate, for the first time, that AA promotes an epithelial-to-mesenchymal-like transition in MCF10A human mammary non-tumorigenic epithelial cells.
Collapse
|
321
|
Tobar N, Villar V, Santibanez JF. ROS-NFkappaB mediates TGF-beta1-induced expression of urokinase-type plasminogen activator, matrix metalloproteinase-9 and cell invasion. Mol Cell Biochem 2010; 340:195-202. [PMID: 20204677 DOI: 10.1007/s11010-010-0418-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 02/10/2010] [Indexed: 12/27/2022]
Abstract
TGF-beta1 has been postulated as a pro-oncogenic factor in the late step of the tumoral progression. In transformed cells, TGF-beta1 enhances the capacity to degrade the extracellular matrix, cell invasiveness and epithelial-mesenchymal transition, which are crucial steps for metastasis. Urokinase-type plasminogen activator (uPA) and matrix metalloproteinase-9 (MMP-9) are critical components in cell migration and invasion induced by TGF-beta1, however, the exact mechanism by which TGF-beta1 regulates uPA and MMP-9 is not well elucidated so far. In the present study, we analyzed the role of ROS-NFkappaB, signal as mediator in the cell malignity enhancement by TGF-beta1. We found that TGF-beta1 activates NFkappaB, through Rac1-NOXs-ROS-dependent mechanism. Our results shows that TGF-beta1 stimulation of uPA and MMP-9 expression involve NOXs-dependent ROS and NFkappaB, activation, demonstrated by using DPI, NOXs inhibitor, ROS scavenger N-acetylcysteine and SN50, an NFkb inhibitor. Furthermore, we found that the inhibition of ROS and NFkappaB, abrogates TGF-beta1 stimulation of EMT, cell motility and invasion. Thus, ROS-NFkappaB acts as the crucial signal in TGF-beta1-induced uPA and MMP-9 expression thereby mediating the enhancement of cellular malignity by TGF-beta1.
Collapse
Affiliation(s)
- Nicolas Tobar
- Laboratorio de Biología Celular, Instituto de Nutrición y Tecnología de los Alimentos, (INTA), Universidad de Chile, Santiago, Chile
| | | | | |
Collapse
|
322
|
Cho KB, Cho MK, Lee WY, Kang KW. Overexpression of c-myc induces epithelial mesenchymal transition in mammary epithelial cells. Cancer Lett 2010; 293:230-9. [PMID: 20144848 DOI: 10.1016/j.canlet.2010.01.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 01/15/2010] [Accepted: 01/16/2010] [Indexed: 01/15/2023]
Abstract
The c-myc gene is frequently overexpressed in human breast cancer and its target genes are involved in tumorigenesis. Epithelial mesenchymal transitions (EMT), where cells undergo a developmental switch from a polarized epithelial phenotype to a highly motile mesenchymal phenotype, are associated with invasion and motility of cancer cells. Basal E-cadherin expression was down-regulated in c-myc overexpressing MCF10A (c-myc-MCF10A) cells compared to GFP-overexpressing MCF10A (GFP-MCF10A) cells, while N-cadherin was distinctly increased in c-myc-MCF10A cells. Given that glycogen synthase kinase-3beta (GSK-3beta) and the snail axis have key roles in E-cadherin deregulation during EMT, we investigated the role of GSK-3beta/snail signaling pathways in the induction of EMT by c-myc overexpression. In contrast to GFP-MCF10A cells, both the transcriptional activity and the ubiquitination-dependent protein stability of snail were enhanced in c-myc-MCF10A cells, and this was reversed by GSK-3beta overexpression. We also found that c-myc overexpression inhibits GSK-3beta activity through activation of extracellular signal-regulated kinase (ERK). Inhibition of ERK by dominant negative mutant transfection or chemical inhibitor significantly suppressed snail gene transcription. These results suggest that c-myc overexpression during transformation of mammary epithelial cells (MEC) is involved in EMTs via ERK-dependent GSK-3beta inactivation and subsequent snail activation.
Collapse
Affiliation(s)
- Kyoung Bin Cho
- BK21 Project Team, College of Pharmacy, Chosun University, Seosuk-dong, Dong-gu, Gwangju 501-759, Republic of Korea
| | | | | | | |
Collapse
|
323
|
Kong D, Li Y, Wang Z, Banerjee S, Ahmad A, Kim HRC, Sarkar FH. miR-200 regulates PDGF-D-mediated epithelial-mesenchymal transition, adhesion, and invasion of prostate cancer cells. Stem Cells 2010; 27:1712-21. [PMID: 19544444 DOI: 10.1002/stem.101] [Citation(s) in RCA: 250] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
MicroRNAs have been implicated in tumor progression. Recent studies have shown that the miR-200 family regulates epithelial-mesenchymal transition (EMT) by targeting zinc-finger E-box binding homeobox 1 (ZEB1) and ZEB2. Emerging evidence from our laboratory and others suggests that the processes of EMT can be triggered by various growth factors, such as transforming growth factor beta and platelet-derived growth factor-D (PDGF-D). Moreover, we recently reported that overexpression of PDGF-D in prostate cancer cells (PC3 PDGF-D cells) leads to the acquisition of the EMT phenotype, and this model offers an opportunity for investigating the molecular interplay between PDGF-D signaling and EMT. Here, we report, for the first time, significant downregulation of the miR-200 family in PC3 PDGF-D cells as well as in PC3 cells exposed to purified active PDGF-D protein, resulting in the upregulation of ZEB1, ZEB2, and Snail2 expression. Interestingly, re-expression of miR-200b in PC3 PDGF-D cells led to reversal of the EMT phenotype, which was associated with the downregulation of ZEB1, ZEB2, and Snail2 expression, and these results were consistent with greater expression levels of epithelial markers. Moreover, transfection of PC3 PDGF-D cells with miR-200b inhibited cell migration and invasion, with concomitant repression of cell adhesion to the culture surface and cell detachment. From these results, we conclude that PDGF-D-induced acquisition of the EMT phenotype in PC3 cells is, in part, a result of repression of miR-200 and that any novel strategy by which miR-200 could be upregulated would become a promising approach for the treatment of invasive prostate cancer.
Collapse
Affiliation(s)
- Dejuan Kong
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | |
Collapse
|
324
|
Pantuck AJ, An J, Liu H, Rettig MB. NF-kappaB-dependent plasticity of the epithelial to mesenchymal transition induced by Von Hippel-Lindau inactivation in renal cell carcinomas. Cancer Res 2010; 70:752-61. [PMID: 20068166 DOI: 10.1158/0008-5472.can-09-2211] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The critical downstream signaling consequences contributing to renal cancer as a result of loss of the tumor suppressor gene von Hippel-Lindau (VHL) have yet to be fully elucidated. Here, we report that VHL loss results in an epithelial to mesenchymal transition (EMT). In studies of paired isogenic cell lines, VHL silencing increased the levels of N-cadherin and vimentin and reduced the levels of E-cadherin relative to the parental VHL(+) cell line, which displayed the opposite profile. VHL(+) cells grew as clusters of cuboidal and rhomboid cells, whereas VHL-silenced cells took on an elongated, fibroblastoid morphology associated with a more highly invasive character in Matrigel chamber assays. Based on earlier evidence that VHL loss can activate NF-kappaB, a known mediator of EMT, we tested whether NF-kappaB contributed to VHL-mediated effects on EMT. On pharmacologic or molecular inhibition of NF-kappaB, VHL-silenced cells regained expression of E-cadherin, lost expression of N-cadherin, and reversed their highly invasive phenotype. Introducing a pVHL-resistant hypoxia-inducible factor 1alpha (HIF1alpha) mutant (HIFalpha(M)) into VHL(+) cells heightened NF-kappaB activity, phenocopying EMT effects produced by VHL silencing. Conversely, inhibiting the heightened NF-kappaB activity in this setting reversed the EMT phenotype. Taken together, these results suggest that VHL loss induces an EMT that is largely dependent on HIFalpha-induced NF-kappaB. Our findings rationalize targeting the NF-kappaB pathway as a therapeutic strategy to treat renal tumors characterized by biallelic VHL inactivation.
Collapse
Affiliation(s)
- Allan J Pantuck
- Department of Urology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095-1738, USA
| | | | | | | |
Collapse
|
325
|
Cane G, Ginouvès A, Marchetti S, Buscà R, Pouysségur J, Berra E, Hofman P, Vouret-Craviari V. HIF-1alpha mediates the induction of IL-8 and VEGF expression on infection with Afa/Dr diffusely adhering E. coli and promotes EMT-like behaviour. Cell Microbiol 2009; 12:640-53. [PMID: 20039880 DOI: 10.1111/j.1462-5822.2009.01422.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Microbes regulate a large panel of intracellular signalling events that can promote inflammation and/or enhance tumour progression. Indeed, it has been shown that infection of human intestinal cells with the Afa/Dr diffusely adhering E. coli C1845 strain induces expression of pro-angiogenic and pro-inflammatory genes. Here, we demonstrate that exposure of cryptic-like intestinal epithelial cells to C1845 bacteria induces HIF-1alpha protein levels. This effect depends on the binding of F1845 adhesin to the membrane-associated DAF receptor that initiates signalling cascades promoting translational mechanisms. Indeed, inhibition of MAPK and PI-3K decreases HIF-1alpha protein levels and blocks C1845-induced phosphorylation of the ribosomal S6 protein. Using RNA interference we show that bacteria-induced HIF-1alpha regulates the expression of IL-8, VEGF and Twist1, thereby pointing to a role for HIF-1 in angiogenesis and inflammation. In addition, infection correlates with a loss of E-cadherin and cytokeratin 18 and a rise in fibronectin, suggesting that bacteria may induce an epithelial to mesenchymal transition-like phenotype. Since HIF-1alpha silencing results in reversion of bacteria-induced EMT markers, we speculate that HIF-1alpha plays a key role linking bacterial infection to angiogenesis, inflammation and some aspects of cancer initiation.
Collapse
|
326
|
Abstract
It has been increasingly recognized that tumor microenvironment plays an important role in carcinogenesis. Inflammatory component is present and contributes to tumor proliferation, angiogenesis, metastasis and resistance to hormonal and chemotherapy. This review highlights the role of inflammation in the tumor metastasis. We focus on the function of proinflammatory factors, particularly cytokines during tumor metastasis. Understanding of the mechanisms by which inflammation contributes to metastasis will lead to innovative approach for treating cancer. How tumor spread remains an enigma and has received great attention in recent years, as metastasis is the major cause of cancer mortality. The complex and highly selective metastatic cascade not only depends on the intrinsic properties of tumor cells but also the microenvironment that they derive from. An inflammatory milieu consisting of infiltrated immune cells and their secretory cytokines, chemokines and growth factors contribute significantly to the invasive and metastatic traits of cancer cells. Here, we review new insights into the molecular pathways that link inflammation in the tumor microenvironment to metastasis.
Collapse
Affiliation(s)
- Yadi Wu
- Department of Molecular and Biomedical Pharmacology, University of Kentucky School of Medicine, Lexington, KY, USA
| | | |
Collapse
|
327
|
Bendinelli P, Matteucci E, Maroni P, Desiderio MA. NF-kappaB activation, dependent on acetylation/deacetylation, contributes to HIF-1 activity and migration of bone metastatic breast carcinoma cells. Mol Cancer Res 2009; 7:1328-41. [PMID: 19671685 DOI: 10.1158/1541-7786.mcr-08-0548] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Here, we show that NF-kappaB-HIF-1 interaction contributed to breast cancer metastatic capacity by means of an incomplete epithelial/mesenchymal transition and influencing migration, as shown in 1833 (human) and 4T1 (mouse) metastatic cells after different stimuli. The 1833 and the transforming growth factor-beta1-exposed 4T1 cells showed both epithelial (E-cadherins) and mesenchymal (N-cadherins and vimentin) markers, and common mechanisms contributed to the retention of certain epithelial characteristics and the control of migration. The complex NF-kappaB-HIF-1 reciprocal regulation and the enhanced c-Jun expression played a functional role in exacerbating the invasiveness of 1833 cells after p50/p65 transfection and of 4T1 cells exposed to transforming growth factor-beta1. Twist expression seemed to exert a permissive role also regulating epithelial/mesenchymal transition markers. After c-Src wild-type (Srcwt) transfection, c-Src-signal transducer overexpression in 1833 cells increased HIF-1 transactivating activity and invasiveness, and changed E-cadherin/N-cadherin ratio versus mesenchymal phenotype. The transcription factor pattern and the motile phenotype of metastatic 1833 cells were influenced by p65-lysine acetylation and HDAC-dependent epigenetic mechanisms, which positively regulated basal NF-kappaB and HIF-1 activities. However, HDAC3 acted as a corepressor of NF-kappaB activity in parental MDA-MB231 cells, thus explaining many differences from the derived 1833 clone, including reduced HIF-1alpha and c-Jun expression. Invasiveness was differently affected by HDAC knockdown in 1833 and MDA-MB231 cells. We suggest that acetylation/deacetylation are critical in establishing the bone-metastatic gene signature of 1833 cells by regulating the activity of NF-kappaB and HIF-1, and further clarify the epigenetic control of transcription factor network in the motile phenotype of 1833 cells.
Collapse
Affiliation(s)
- Paola Bendinelli
- Dipartimento di Morfologia Umana e Scienze Biomediche Città Studi, University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
328
|
Truncation of the catalytic domain of the cylindromatosis tumor suppressor impairs lung maturation. Neoplasia 2009; 11:469-76. [PMID: 19412431 DOI: 10.1593/neo.81424] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 02/24/2009] [Accepted: 02/25/2009] [Indexed: 01/08/2023] Open
Abstract
Cyld encodes a 956-amino acid deubiquitinating enzyme (CYLD), which is a negative regulator of nuclear factor kappaB and mitogen-activated protein kinase pathways. Mutations that truncate and inactivate the carboxyl-terminal deubiquitinating domain of CYLD underlie the development of skin appendage tumors in humans, whereas down-regulation of Cyld expression has been associated with the development of various types of human malignancies including lung cancer. To establish an animal model of human CYLD inactivation and characterize the biological role of CYLD in vivo, we generated mice carrying a homozygous deletion of Cyld exon 9 (Cyld(Delta 9/Delta 9) mice) using a conditional approach. Deletion of exon 9 would cause a carboxyl-terminal truncation of CYLD and inactivation of its deubiquitinating activity. In accordance with previous studies, fibroblasts from Cyld(Delta 9/Delta 9) embryos had hyperactive nuclear factor kappaB and c-Jun kinase pathways compared with control fibroblasts. Cyld(Delta 9/Delta 9) newborn mice were smaller than wild-type littermates with a short and kinky tail and no major developmental defects. However, Cyld(Delta 9/Delta 9) mice died shortly after birth from apparent respiratory dysfunction. Histological examination of E18.5 Cyld(Delta 9/Delta 9) lungs demonstrated an immature phenotype characterized by hyperplasic mesenchyme but apparently normal epithelial, smooth muscle. and endothelial structures. Our study identifies an important role of CYLD in lung maturation, which may underlie the development of many cases of lung cancer.
Collapse
|
329
|
Yoshida S, Furukawa N, Haruta S, Tanase Y, Kanayama S, Noguchi T, Sakata M, Yamada Y, Oi H, Kobayashi H. Expression Profiles of Genes Involved in Poor Prognosis of Epithelial Ovarian Carcinoma: A Review. Int J Gynecol Cancer 2009; 19:992-7. [DOI: 10.1111/igc.0b013e3181aaa93a] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background:Epithelial ovarian cancer (EOC) is the commonest cause of gynecological cancer-related mortality. Although the prognosis for patients with advanced cancer is poor, there is a wide range of outcomes for individual patients.Objective:The aim of this study was to review molecular factors predictive of poor prognosis of women with EOC by reviewing microarray research identifying gene expression profiles.Methods:A systematic search was performed in the electronic databases PubMed and ScienceDirect up to July 2008, combining the keywords "genome-wide," "microarray," "epithelial ovarian cancer" "prognosis," and "epithelial-mesenchymal transition" with specific expression profiles of genes.Results:Many genes that participated in cell signaling, growth factors, transcription factors, proteinases, metabolism, cell adhesion, extracellular matrix component, cell proliferation, and anti-apoptosis were overexpressed in patients with poor prognosis. Several important prognosis-related genes overlap with those known to be regulated by epithelial-mesenchymal transition (EMT). This signaling pathway of EMT (E-cadherin, β-catenin, receptor tyrosine kinases, NF-κB, TGF-β, or Wnt signalings) will be discussed, as it provides new insights into a new treatment strategy.Conclusions:This review summarizes recent advances in prognosis-related molecular biology. Collectively, molecular changes possibly through EMT are considered to be a major contributor to the poor prognosis of EOC.
Collapse
|
330
|
Valacchi G, Pecorelli A, Mencarelli M, Carbotti P, Fortino V, Muscettola M, Maioli E. Rottlerin: a multifaced regulator of keratinocyte cell cycle. Exp Dermatol 2009; 18:516-21. [DOI: 10.1111/j.1600-0625.2008.00816.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
331
|
Dominguez I, Sonenshein GE, Seldin DC. Protein kinase CK2 in health and disease: CK2 and its role in Wnt and NF-kappaB signaling: linking development and cancer. Cell Mol Life Sci 2009; 66:1850-7. [PMID: 19387549 PMCID: PMC3905806 DOI: 10.1007/s00018-009-9153-z] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
CK2 is a highly conserved tetrameric serine/ threonine kinase present in all eukaryotic organisms. It is constitutively active, and appears to be regulated by level of expression and activity, and subcellular localization. In turn, it has been postulated to control the function of many proteins through changes in phosphorylation that affect protein stability, protein-protein interactions, and subcellular localization. Through these mechanisms, CK2 regulates many fundamental cellular properties. An enzyme that carries out such a master regulatory function is likely to be important in organismic development and in cancer. We have shown that overexpression of CK2 catalytic subunits is capable of promoting tumorigenesis, and that loss of CK2 catalytic subunits in development can be lethal. Through studies in cells, mice, and frogs, we and others have identified the Wnt and NF-kappaB pathways as two key signal transduction pathways that are regulated by CK2 activity, in embryonic development and in cancer. These results suggest that inhibiting CK2 could be useful in treating cancer, but dangerous to developing organisms.
Collapse
Affiliation(s)
- I. Dominguez
- Section of Hematology-Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, 650 Albany Street, Boston, Massachusetts 02118 USA
| | - G. E. Sonenshein
- Section of Hematology-Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, 650 Albany Street, Boston, Massachusetts 02118 USA
| | - D. C. Seldin
- Section of Hematology-Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, 650 Albany Street, Boston, Massachusetts 02118 USA
| |
Collapse
|
332
|
Jiang P, Enomoto A, Takahashi M. Cell biology of the movement of breast cancer cells: intracellular signalling and the actin cytoskeleton. Cancer Lett 2009; 284:122-30. [PMID: 19303207 DOI: 10.1016/j.canlet.2009.02.034] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 02/04/2009] [Accepted: 02/16/2009] [Indexed: 12/16/2022]
Abstract
Cell motility is a critical step in cancer invasion and metastasis that must be unravelled to gain an appropriate understanding of the behaviour of cancer cells. A broad spectrum of motility mechanisms that facilitate invasion of extramammary tissues and metastasis exists in breast cancer cells (e.g. reorganization of the actin cytoskeleton, regulation of focal adhesion, changes in response to a different microenvironment, epithelial mesenchymal transition, and control of membrane proteins through endocytosis). These cellular responses are tightly regulated by intracellular signalling pathways evoked by humoral factors that include growth factors, chemokines, and cytokines. Learning more about the cellular and molecular basis of these different motility programmes will aid in the development of treatments for breast cancer invasion and metastasis. This review of recent literature focuses on aspects of cell biology related to motility and metastasis, and suggests some directions for future breast cancer research.
Collapse
Affiliation(s)
- Ping Jiang
- Department of Pathology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | | | | |
Collapse
|
333
|
Wang Z, Li Y, Kong D, Banerjee S, Ahmad A, Azmi AS, Ali S, Abbruzzese JL, Gallick GE, Sarkar FH. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res 2009; 69:2400-7. [PMID: 19276344 DOI: 10.1158/0008-5472.can-08-4312] [Citation(s) in RCA: 526] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite rapid advances in many fronts, pancreatic cancer (PC) remains one of the most difficult human malignancies to treat due, in part, to de novo and acquired chemoresistance and radioresistance. Gemcitabine alone or in combination with other conventional therapeutics is the standard of care for the treatment of advanced PC without any significant improvement in the overall survival of patients diagnosed with this deadly disease. Previous studies have shown that PC cells that are gemcitabine-resistant (GR) acquired epithelial-mesenchymal transition (EMT) phenotype, which is reminiscent of "cancer stem-like cells"; however, the molecular mechanism that led to EMT phenotype has not been fully investigated. The present study shows that Notch-2 and its ligand, Jagged-1, are highly up-regulated in GR cells, which is consistent with the role of the Notch signaling pathway in the acquisition of EMT and cancer stem-like cell phenotype. We also found that the down-regulation of Notch signaling was associated with decreased invasive behavior of GR cells. Moreover, down-regulation of Notch signaling by siRNA approach led to partial reversal of the EMT phenotype, resulting in the mesenchymal-epithelial transition, which was associated with decreased expression of vimentin, ZEB1, Slug, Snail, and nuclear factor-kappaB. These results provide molecular evidence showing that the activation of Notch signaling is mechanistically linked with chemoresistance phenotype (EMT phenotype) of PC cells, suggesting that the inactivation of Notch signaling by novel strategies could be a potential targeted therapeutic approach for overcoming chemoresistance toward the prevention of tumor progression and/or treatment of metastatic PC.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
334
|
Hahn ME, Allan LL, Sherr DH. Regulation of constitutive and inducible AHR signaling: complex interactions involving the AHR repressor. Biochem Pharmacol 2008; 77:485-97. [PMID: 18848529 DOI: 10.1016/j.bcp.2008.09.016] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 09/10/2008] [Accepted: 09/11/2008] [Indexed: 01/13/2023]
Abstract
The AHR is well known for regulating responses to an array of environmental chemicals. A growing body of evidence supports the hypothesis that the AHR also plays perhaps an even more important role in modulating critical aspects of cell function including cell growth, death, and migration. As these and other important AHR activities continue to be elucidated, it becomes apparent that attention now must be directed towards the mechanisms through which the AHR itself is regulated. Here, we review what is known of and what biological outcomes have been attributed to the AHR repressor (AHRR), an evolutionarily conserved bHLH-PAS protein that inhibits both xenobiotic-induced and constitutively active AHR transcriptional activity in multiple species. We discuss the structure and evolution of the AHRR and the dominant paradigm of a xenobiotic-inducible negative feedback loop comprised of AHR-mediated transcriptional up-regulation of AHRR and the subsequent AHRR-mediated suppression of AHR activity. We highlight the role of the AHRR in limiting AHR activity in the absence of xenobiotic AHR ligands and the important contribution of constitutively repressive AHRR to cancer biology. In this context, we also suggest a new hypothesis proposing that, under some circumstances, constitutively active AHR may repress AHRR transcription, resulting in unbridled AHR activity. We also review the predominant hypotheses on the molecular mechanisms through which AHRR inhibits AHR as well as novel mechanisms through which the AHRR may exert AHR-independent effects. Collectively, this discussion emphasizes the importance of this understudied bHLH-PAS protein in tissue development, normal cell biology, xenobiotic responsiveness, and AHR-regulated malignancy.
Collapse
Affiliation(s)
- Mark E Hahn
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | | | | |
Collapse
|