Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neoplasia ; 35: 100855, 2023 01.
Article in English | MEDLINE | ID: mdl-36410227

ABSTRACT

Eosinophils are terminally differentiated leukocytes that participate in the process of chronic inflammation and allergy and are able to release multiple cytokines into the surrounding tissue environment. Tumor-associated tissue eosinophilia (TATE) is the presence of eosinophils in the tumor or in the neighboring stroma and has been observed in various types of cancer. In head and neck squamous cell carcinoma (HNSCC), the clinical relevance of TATE has not been concluded yet because of the inconsistent results in different studies. In our study, we focus on the prognostic effects of TATE on HNSCC and how TATE can influence tumor behavior and tumor microenvironment. We first showed that in both the TCGA-HNSC cohort and our cohort of patients with HNSCC who had received curative surgery, TATE is correlated with worse overall survival. To investigate the underlying mechanism of how TATE leads to poor clinical outcomes, we showed that activated eosinophils produce a variety of cytokines and chemokines, and activated TATE-derived culture medium promotes tumor migration mainly through CCL2. We also showed that eosinophils are capable of inducing angiogenesis and that HNSCC samples enriched with TATE are highly correlated with tumor angiogenesis. Furthermore, HNSCC enriched with TATE had more aggressive pathological features, including regional lymph node metastasis, perineural invasion, lymphovascular invasion, and tumor growth. Lastly, we showed that HNSCC enriched with TATE is associated with immunosuppressive tumor microenvironment. Taken together, our results suggest that TATE promotes cancer metastasis and angiogenesis which results in a poor clinical outcomes in HNSCC.


Subject(s)
Carcinoma, Squamous Cell , Eosinophilia , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck , Carcinoma, Squamous Cell/pathology , Eosinophilia/etiology , Eosinophilia/pathology , Prognosis , Cytokines , Tumor Microenvironment
2.
Cancers (Basel) ; 13(8)2021 Apr 12.
Article in English | MEDLINE | ID: mdl-33921319

ABSTRACT

Cell migration is critical for regional dissemination and distal metastasis of cancer cells, which remain the major causes of poor prognosis and death in patients with colorectal cancer (CRC). Although cytoskeletal dynamics and cellular deformability contribute to the migration of cancer cells and metastasis, the mechanisms governing the migratory ability of cancer stem cells (CSCs), a nongenetic source of tumor heterogeneity, are unclear. Here, we expanded colorectal CSCs (CRCSCs) as colonospheres and showed that CRCSCs exhibited higher cell motility in transwell migration assays and 3D invasion assays and greater deformability in particle tracking microrheology than did their parental CRC cells. Mechanistically, in CRCSCs, microRNA-210-3p (miR-210) targeted stathmin1 (STMN1), which is known for inducing microtubule destabilization, to decrease cell elasticity in order to facilitate cell motility without affecting the epithelial-mesenchymal transition (EMT) status. Clinically, the miR-210-STMN1 axis was activated in CRC patients with liver metastasis and correlated with a worse clinical outcome. This study elucidates a miRNA-oriented mechanism regulating the deformability of CRCSCs beyond the EMT process.

3.
Biochem Biophys Res Commun ; 522(4): 1009-1014, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31813546

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α) induces cancer metastasis. We previously demonstrated that HIF-1α-induced membrane-type 4 matrix metalloproteinase (MT4-MMP) is involved in hypoxia-mediated metastasis in head and neck squamous cell carcinoma (HNSCC). However, the functions and detailed mechanisms of MT4-MMP in cancer metastasis are not well understood. In this study, we investigated whether MT4-MMP regulates invadopodia formation or individual cell movement-both critical to cancer migration and invasion-in three-dimensional (3D) environments. By expressing MT4-MMP in the HNSCC cell line FaDu, we demonstrated that MT4-MMP increases invadopodia formation and gelatin degradation. Furthermore, the amoeboid-like cell movement on collagen gel was increased by MT4-MMP expression in FaDu cells. Mechanistically, MT4-MMP may induce invadopodia formation by binding with Tks5 and PDGFRα to result in Src activation and promote amoeboid-like movement by stimulating the small GTPases Rho and Cdc42. Altogether, our data indicate that MT4-MMP induces two crucial mechanisms of cancer dissemination, invadopodia formation and amoeboid movement, and elucidate the prometastatic role of MT4-MMP in hypoxia-mediated cancer metastasis.


Subject(s)
Cell Movement , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/pathology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Podosomes/pathology , Adaptor Proteins, Vesicular Transport/metabolism , Cardiac Myosins/metabolism , Cell Line, Tumor , Gelatin/metabolism , HEK293 Cells , Humans , Myosin Light Chains/metabolism , Neoplasm Invasiveness , Phosphorylation , Phosphotyrosine/metabolism , Protein Binding , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , src-Family Kinases/metabolism
4.
Int J Cancer ; 145(8): 2209-2224, 2019 10 15.
Article in English | MEDLINE | ID: mdl-30980673

ABSTRACT

The dynamic cell-cell communication is essential for tissue homeostasis in normal physiological circumstances and contributes to a diversified tumor microenvironment. Although exosomes are extracellular vesicles that actively participate in cell-cell interaction by shutting cellular components, impacts of tumor exosomes in the context of cancer stemness remain elusive. Here, we expand colorectal cancer stem cells (CRCSCs) as cancer spheroids and demonstrate that the ß-catenin/Tcf-4-activated RAB27B expression is required for the secretion of CRCSC exosomes. In an exosomal RNA sequencing analysis, a switch of exosomal RNA species from retrotransposons to microRNAs (miRNAs) is identified upon expanding CRCSCs. miRNA-146a-5p (miR-146a) is the major miRNA in CRCSC exosomes and exosomal miR-146a promotes stem-like properties and tumorigenicity by targeting Numb in recipient CRC cells. Among 53 CRC patients, those with abundant exosomal miR-146a expression in serum exhibits higher miR-146aHigh /NumbLow CRCSC traits, an increased number of tumor-filtrating CD66(+) neutrophils and a decreased number of tumor-infiltrating CD8(+) T cells. Our study elucidates a unique mechanism of tumor exosome-mediated stemness expansion.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Exosomes/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , rab GTP-Binding Proteins/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , HT29 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA Interference , Tumor Microenvironment/genetics , rab GTP-Binding Proteins/metabolism
5.
Nat Cell Biol ; 21(4): 533, 2019 04.
Article in English | MEDLINE | ID: mdl-30833696

ABSTRACT

In the version of Supplementary Fig. 3c originally published with this Article, the authors mistakenly duplicated a blot from Supplementary Fig. 3b. The correct versions of these figures are shown below. In addition, two independent repeats of the experiments presented in Supplementary Figs. 3b and 3c, showing results consistent with those originally reported, have been deposited in Figshare ( 10.6084/m9.figshare.7545263 ).

6.
Nat Cell Biol ; 21(5): 664, 2019 05.
Article in English | MEDLINE | ID: mdl-30820041

ABSTRACT

In the version of Supplementary Fig. 6c originally published with this Article, the immunoprecipitation (IP) and immunoblotting (IB) tags in the top panel were mislabelled. In addition, in Supplementary Fig. 6e, the blot of the IP: Numb; IB: ß-Trcp panel for HCT15 was mistakenly duplicated for HCT116. The correct versions of these figures are shown below. An independent repeat of the experiments presented in Supplementary Fig. 6c and e, showing results that are consistent with those reported in the unprocessed blots, have been deposited in figshare ( 10.6084/m9.figshare.7570685 ).

7.
J Hematol Oncol ; 12(1): 10, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30683126

ABSTRACT

BACKGROUND: Cell-cell interactions maintain tissue homeostasis and contribute to dynamic alteration of the tumor microenvironment (TME). Communication between cancer and host cells not only promotes advanced disease aggression but also determines therapeutic response in cancer patients. Despite accumulating evidence supporting the role of tumor-infiltrating immunocytes in modulating tumor immunity, the interplay between heterogeneous tumor subpopulations and immunocytes is elusive. METHODS: We expanded colorectal cancer stem cells (CRCSCs) as cancer spheroids from the murine colorectal cancer (CRC) cell line CT26 to interrogate tumor-host interactions using a syngeneic tumor model. RNA-sequencing analysis of host cells and tumor exosomes was performed to identify molecular determinants that mediate the crosstalk between CRCSCs and immunocytes. The Cancer Genome Atlas (TCGA) database was used to validate the clinical significance in CRC patients. RESULTS: The expanded CT26 cancer spheroids showed increased stemness gene expression, enhanced spheroid and clonogenicity potential, and an elevated tumor-initiating ability, characteristic of CRCSCs. By examining immune cell composition in syngeneic tumor-bearing mice, a systemic increase in CD11b+/Ly6GHigh/Ly6CLow neutrophils was observed in mice bearing CRCSC-derived tumors. An increased secretion of CRCSC exosomes was observed in vitro, and through in vivo tracking, CRCSC exosomes were found to be transported to the bone marrow. Moreover, CRCSC exosomes prolonged the survival of bone marrow-derived neutrophils and engendered a protumoral phenotype in neutrophils. Mechanistically, tumor exosomal tri-phosphate RNAs induced the expression of interleukin-1ß (IL-1ß) through a pattern recognition-NF-κB signaling axis to sustain neutrophil survival. CRCSC-secreted CXCL1 and CXCL2 then attracted CRCSC-primed neutrophils to promote tumorigenesis of CRC cells via IL-1ß. Moreover, neutrophil depletion using a Ly6G-specific antibody (clone 1A8) attenuated the tumorigenicity of CRCSCs. In human specimens, CRC patients exhibiting an active CRCSC signal (Snail+IL8+) showed elevated tumor infiltration of MPO+ neutrophils, and high (in the top 10%) MPO expression predicted poor survival of CRC patients. CONCLUSIONS: This study elucidates a multistep CRCSC-neutrophil interaction during advanced cancer progression. Strategies targeting aberrant neutrophil activation may be developed for combating CSC-related malignancy.


Subject(s)
Carcinogenesis/metabolism , Cell Communication , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Exosomes/metabolism , Neoplastic Stem Cells/metabolism , Neutrophils/metabolism , RNA/genetics , Animals , Base Sequence , Cell Line, Tumor , Cell Survival , HEK293 Cells , Humans , Interleukin-1beta/metabolism , Isografts , Mice , Mice, Inbred BALB C , NF-kappa B p50 Subunit/metabolism , Peroxidase/metabolism , Spheroids, Cellular/metabolism , Tumor Microenvironment
8.
Nat Cell Biol ; 21(2): 251-262, 2019 02.
Article in English | MEDLINE | ID: mdl-30664792

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a pivotal mechanism for cancer dissemination. However, EMT-regulated individual cancer cell invasion is difficult to detect in clinical samples. Emerging evidence implies that EMT is correlated to collective cell migration and invasion with unknown mechanisms. We show that the EMT transcription factor Snail elicits collective migration in squamous cell carcinoma by inducing the expression of a tight junctional protein, claudin-11. Mechanistically, tyrosine-phosphorylated claudin-11 activates Src, which suppresses RhoA activity at intercellular junctions through p190RhoGAP, maintaining stable cell-cell contacts. In head and neck cancer patients, the Snail-claudin-11 axis prompts the formation of circulating tumour cell clusters, which correlate with tumour progression. Overexpression of snail correlates with increased claudin-11, and both are associated with a worse outcome. This finding extends the current understanding of EMT-mediated cellular migration via a non-individual type of movement to prompt cancer progression.


Subject(s)
Cell Movement/genetics , Claudins/genetics , Neoplasms/genetics , Snail Family Transcription Factors/genetics , Animals , Caco-2 Cells , Cell Line, Tumor , Cells, Cultured , Claudins/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Snail Family Transcription Factors/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
9.
J Cell Mol Med ; 22(8): 3837-3846, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29726584

ABSTRACT

The biological impact and signalling of epithelial-mesenchymal transition (EMT) during cancer metastasis has been established. However, the changes in biophysical properties of cancer cells undergoing EMT remain elusive. Here, we measured, via video particle tracking microrheology, the intracellular stiffness of head and neck cancer cell lines with distinct EMT phenotypes. We also examined cells migration and invasiveness in different extracellular matrix architectures and EMT-related signalling in these cell lines. Our results show that when cells were cultivated in three-dimensional (3D) environments, the differences in cell morphology, migration speed, invasion capability and intracellular stiffness were more pronounced among different head and neck cancer cell lines with distinct EMT phenotypes than those cultivated in traditional plastic dishes and/or seated on top of a thick layer of collagen. An inverse correlation between intracellular stiffness and invasiveness in 3D culture was revealed. Knock-down of the EMT regulator Twist1 or Snail or inhibition of Rac1 which is a downstream GTPase of Twist1 increased intracellular stiffness. These results indicate that the EMT regulators, Twist1 and Snail and the mediated signals play a critical role in reducing intracellular stiffness and enhancing cell migration in EMT to promote cancer cells invasion.

10.
Clin Cancer Res ; 23(15): 4388-4401, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28196873

ABSTRACT

Purpose: In head and neck squamous cell carcinoma (HNSCC), the incidence of RAS mutation, which is the major cause of cetuximab resistance, is relatively rare compared with the other types of cancers, and the mechanism mediating acquired resistance is unclear compared with the driver gene mutation-mediated de novo resistance. Here, we investigated the driver gene mutation-independent mechanism for cetuximab resistance in HNSCC.Experimental Design: We used the in vitro-selected and in vivo-selected cetuximab-resistant sublines of HNSCC cell lines for investigating the mechanism of acquired resistance to cetuximab. Zebrafish model was applied for evaluating the synergistic effect of combinatory drugs for overcoming cetuximab resistance.Results: The cetuximab-resistant HNSCC cells undergo a Snail-induced epithelial-mesenchymal transition. Mechanistically, Snail induces the expression of lymphotoxin-ß (LTß), a TNF superfamily protein that activates NF-κB, and protein arginine methyltransferase 1 (PRMT1), an arginine methyltransferase that methylates EGFR. LTß interacts with methylated EGFR to promote its ligand-binding ability and dimerization. Furthermore, LTß activates the NF-κB pathway through a LTß receptor-independent mechanism. Combination of an EGFR tyrosine kinase inhibitor and a NF-κB inhibitor effectively suppressed cetuximab-resistant HNSCC and interfering with the EGFR-LTß interaction reverses resistance.Conclusions: Our findings elucidate the mechanism of driver gene mutations-independent mechanism of acquired resistance to cetuximab in HNSCC and also provide potential strategies for combating cetuximab resistance. Clin Cancer Res; 23(15); 4388-401. ©2017 AACR.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Epithelial-Mesenchymal Transition/genetics , ErbB Receptors/genetics , Head and Neck Neoplasms/drug therapy , Lymphotoxin-beta/genetics , NF-kappa B/antagonists & inhibitors , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cetuximab/administration & dosage , Cetuximab/adverse effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Mice , Mutation , NF-kappa B/genetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Snail Family Transcription Factors/genetics , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays , Zebrafish , Zebrafish Proteins/genetics
11.
Cell Rep ; 14(3): 520-533, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26776511

ABSTRACT

Let-7 is crucial for both stem cell differentiation and tumor suppression. Here, we demonstrate a chromatin-dependent mechanism of let-7 in regulating target gene expression in cancer cells. Let-7 directly represses the expression of AT-rich interacting domain 3B (ARID3B), ARID3A, and importin-9. In the absence of let-7, importin-9 facilitates the nuclear import of ARID3A, which then forms a complex with ARID3B. The nuclear ARID3B complex recruits histone demethylase 4C to reduce histone 3 lysine 9 trimethylation and promotes the transcription of stemness factors. Functionally, expression of ARID3B is critical for the tumor initiation in let-7-depleted cancer cells. An inverse association between let-7 and ARID3A/ARID3B and prognostic significance is demonstrated in head and neck cancer patients. These results highlight a chromatin-dependent mechanism where let-7 regulates cancer stemness through ARID3B.


Subject(s)
DNA-Binding Proteins/metabolism , MicroRNAs/metabolism , Animals , Base Sequence , Cell Line , Chromatin/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Female , Histones/metabolism , Humans , Karyopherins/chemistry , Karyopherins/genetics , Karyopherins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Octamer Transcription Factor-3/genetics , Oligonucleotides, Antisense/metabolism , RNA Interference , Sequence Alignment , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome , Transplantation, Heterologous
12.
J Cell Mol Med ; 18(7): 1358-71, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24758741

ABSTRACT

Adenomyosis is an oestrogen-dependent disease characterized by the invasion of endometrial epithelial cells into the myometrium of uterus, and angiogenesis is thought to be required for the implantation of endometrial glandular tissues during the adenomyotic pathogenesis. In this study, we demonstrate that compared with eutopic endometria, adenomyotic lesions exhibited increased vascularity as detected by sonography. Microscopically, the lesions also exhibited an oestrogen-associated elevation of microvascular density and VEGF expression in endometrial epithelial cells. We previously reported that oestrogen-induced Slug expression was critical for endometrial epithelial-mesenchymal transition and development of adenomyosis. Our present studies demonstrated that estradiol (E2) elicited a Slug-VEGF axis in endometrial epithelial cells, and also induced pro-angiogenic activity in vascular endothelial cells. The antagonizing agents against E2 or VEGF suppressed endothelial cells migration and tubal formation. Animal experiments furthermore confirmed that blockage of E2 or VEGF was efficient to attenuate the implantation of adenomyotic lesions. These results highlight the importance of oestrogen-induced angiogenesis in adenomyosis development and provide a potential strategy for treating adenomyosis through intercepting the E2-Slug-VEGF pathway.


Subject(s)
Adenomyosis/pathology , Epithelial Cells/pathology , Estrogens/adverse effects , Neovascularization, Pathologic/pathology , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adenomyosis/drug therapy , Adenomyosis/etiology , Animals , Blotting, Western , Cells, Cultured , Endometriosis/drug therapy , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Myometrium/drug effects , Myometrium/metabolism , Myometrium/pathology , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/metabolism , Snail Family Transcription Factors
13.
Nat Cell Biol ; 16(3): 268-80, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24561623

ABSTRACT

Asymmetrical cell division (ACD) maintains the proper number of stem cells to ensure self-renewal. In cancer cells, the deregulation of ACD disrupts the homeostasis of the stem cell pool and promotes tumour growth. However, this mechanism is unclear. Here, we show a reduction of ACD in spheroid-derived colorectal cancer stem cells (CRCSCs) compared with differentiated cancer cells. The epithelial-mesenchymal transition (EMT) inducer Snail is responsible for the ACD-to-symmetrical cell division (SCD) switch in CRCSCs. Mechanistically, Snail induces the expression of microRNA-146a (miR-146a) through the ß-catenin-TCF4 complex. miR-146a targets Numb to stabilize ß-catenin, which forms a feedback circuit to maintain Wnt activity and directs SCD. Interference with the Snail-miR-146a­ß-catenin loop by inhibiting the MEK or Wnt activity reduces the symmetrical division of CRCSCs and attenuates tumorigenicity. In colorectal cancer patients, the Snail(High)Numb(Low) profile is correlated with cetuximab resistance and a poorer prognosis. This study elucidates a unique mechanism of EMT-induced CRCSC expansion.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , MicroRNAs/genetics , Mitosis , Neoplastic Stem Cells/physiology , Transcription Factors/physiology , 3' Untranslated Regions , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Binding Sites , Cell Line, Tumor , Cetuximab , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prognosis , Proteolysis , Snail Family Transcription Factors , Transcription Factor 4 , Transcription Factors/metabolism , Transcription, Genetic , Tumor Burden/drug effects , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/metabolism
14.
Biochem Biophys Res Commun ; 433(1): 24-30, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23454123

ABSTRACT

The movement modes of epithelial cancer cells in three-dimensional (3D) environments include the mesenchymal mode, which is associated with local invasion, and the amoeboid mode, which facilitates distant metastasis. The migratory behavior of individual cancer cells is critical for tumor dissemination; however, the mechanism underlying regulation of the switch between movement modes is not clearly understood. For head and neck squamous cell carcinoma (HNSCC), local invasion is the major route of dissemination. We previously demonstrated that, in HNSCC cells, Twist1 represses let-7i expression to elicit mesenchymal-mode movement through activation of Ras-related C3 botulinum toxin substrate 1 (RAC1). In this study, we discover another important target gene of let-7i for regulating HNSCC migration. Using bioinformatic tools, we identified bone morphogenetic protein 4 (BMP4) as a candidate target of let-7i. Further experiments, including 3'-untranslated region (UTR) reporter assays, quantitative RT-PCR and western blotting, confirmed that BMP4 is a bona fide target repressed by let-7i. In the HNSCC cell line OECM-1, knockdown of BMP4 reduced mesenchymal-mode migration and invasion in 3D culture. In clinical HNSCC samples, let-7i expression was inversely correlated with BMP4 expression. Our results revealed that let-7i attenuates mesenchymal-mode migration of HNSCC cells through repression of a novel target, BMP4.


Subject(s)
Bone Morphogenetic Protein 4/antagonists & inhibitors , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , MicroRNAs/metabolism , 3' Untranslated Regions , Base Sequence , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/secondary , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Head and Neck Neoplasms/genetics , Humans , Mesoderm/metabolism , Mesoderm/pathology , MicroRNAs/genetics , Neoplasm Invasiveness , RNA, Small Interfering/genetics , Signal Transduction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
15.
Nat Cell Biol ; 14(4): 366-74, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22407364

ABSTRACT

Epithelial-mesenchymal transition (EMT), which is characterized by the suppression of the adhesion protein E-cadherin, is a crucial process that promotes metastasis and stem-like properties of cancer cells. However, the dissociation of cellular aggregates is not sufficient to explain why cancer cells move, and the motile nature of cancer cells undergoing EMT remains elusive. Here, we identify a mechanism in which the EMT inducer Twist1 elicits cancer cell movement through activation of RAC1. Twist1 cooperates with BMI1 to suppress let-7i expression, which results in upregulation of NEDD9 and DOCK3, leading to RAC1 activation and enabling mesenchymal-mode movement in three-dimensional environments. Moreover, the suppression of let-7i contributes to Twist1-induced stem-like properties. Clinically, activation of the Twist1-let-7i-NEDD9 axis in head and neck cancer patients correlates with tumour invasiveness and worse outcome. Our results uncover an essential mechanism to explain how Twist1 induces the motile stem-like cancer cell phenotype beyond simply suppressing E-cadherin.


Subject(s)
Cell Movement , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/metabolism , Twist-Related Protein 1/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Line, Tumor , HEK293 Cells , Humans
16.
Gastroenterology ; 141(1): 279-91, 291.e1-5, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21640118

ABSTRACT

BACKGROUND & AIMS: Some cancer cells have activities that are similar to those of stem cells from normal tissues, and cell dedifferentiation correlates with poor prognosis. Little is known about the mechanisms that regulate the stem cell-like features of cancer cells; we investigated genes associated with stem cell-like features of colorectal cancer (CRC) cells. METHODS: We isolated colonospheres from primary CRC tissues and cell lines and characterized their gene expression patterns by microarray analysis. We also investigated the biological features of the colonosphere cells. RESULTS: Expanded CRC colonospheres contained cells that expressed high levels of CD44 and CD166, which are markers of colon cancer stem cells, and had many features of cancer stem cells, including chemoresistance and radioresistance, the ability to initiate tumor formation, and activation of epithelial-mesenchymal transition (EMT). SNAIL, an activator of EMT, was expressed at high levels by CRC colonospheres. Overexpression of Snail in CRC cells induced most properties of colonospheres, including cell dedifferentiation. Two hundred twenty-seven SNAIL-activated genes were up-regulated in colonospheres; gene regulatory networks centered around interleukin (IL)-8 and JUN. Blocking IL-8 expression or activity disrupted SNAIL-induced stem cell-like features of colonospheres. We observed that SNAIL activated the expression of IL8 by direct binding to its E3/E4 E-boxes. In CRC tissues, SNAIL and IL-8 were coexpressed with the stem cell marker CD44 but not with CD133 or CD24. CONCLUSIONS: In human CRC tissues, SNAIL regulates expression of IL-8 and other genes to induce cancer stem cell activities. Strategies that disrupt this pathway might be developed to block tumor formation by cancer stem cells.


Subject(s)
Carcinoma/metabolism , Colorectal Neoplasms/metabolism , Interleukin-8/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Transcription Factors/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibodies/pharmacology , Antigens, CD/metabolism , Antimetabolites, Antineoplastic/pharmacology , Binding Sites , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/immunology , Carcinoma/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Resistance, Neoplasm , E-Box Elements , Epithelial-Mesenchymal Transition/drug effects , Fetal Proteins/metabolism , Fluorouracil/pharmacology , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Hyaluronan Receptors/metabolism , Interleukin-8/genetics , Interleukin-8/immunology , Mice , Mice, Nude , Middle Aged , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Oligonucleotide Array Sequence Analysis , Radiation Tolerance , Signal Transduction/drug effects , Signal Transduction/radiation effects , Snail Family Transcription Factors , Spheroids, Cellular , Time Factors , Transcription Factors/genetics , Transfection , Tumor Burden/drug effects , Tumor Cells, Cultured
17.
Clin Cancer Res ; 16(18): 4561-71, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20823140

ABSTRACT

PURPOSE: We investigated the mechanism and clinical significance of the epithelial-mesenchymal transition (EMT)-induced chemoresistance in head and neck squamous cell carcinoma (HNSCC). EXPERIMENTAL DESIGN: The correlation between the expression of different EMT regulators and chemoresistance genes, such as excision repair cross complementation group 1 (ERCC1), was evaluated in cancer cell lines from the NCI-60 database and four human HNSCC cell lines. Ectopic expression of Snail or short-interference RNA-mediated repression of Snail or ERCC1 was done in HNSCC cell lines. Cell viability was examined for cells after cisplatin treatment. A luciferase reporter assay and chromatin immunoprecipitation were used to identify the transcriptional regulation of ERCC1 by Snail. Immunohistochemical analysis of Snail, Twist1, ERCC1, hypoxia inducible factor-1 α (HIF-1α), and NBS1 were done in samples from 72 HNSCC patients receiving cisplatin-based chemotherapy. RESULTS: The correlation between the expression of Snail and ERCC1 was confirmed in different cell lines, including HNSCC cells. In HNSCC cell lines, overexpression of Snail in the low endogenous Snail/ERCC1 cell lines FaDu or CAL-27 increased ERCC1 expression, and hypoxia or overexpression of NBS1 also upregulated ERCC1. Knockdown of Snail in the high endogenous Snail/ERCC1 cell line OECM-1 downregulated ERCC1 expression and attenuated cisplatin resistance. Furthermore, suppression of ERCC1 in Snail- or NBS1-overexpressing HNSCC cells enhanced sensitivity to cisplatin. Snail directly regulated ERCC1 transcription. In patients with HNSCC, coexpression of Snail and ERCC1 correlated with cisplatin resistance and a poor prognosis. CONCLUSIONS: Activation of ERCC1 by Snail is critical in the generation of cisplatin resistance of HNSCC cells.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cisplatin/therapeutic use , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Endonucleases/genetics , Head and Neck Neoplasms/genetics , Transcription Factors/physiology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/radiotherapy , Humans , Microarray Analysis , Snail Family Transcription Factors , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
18.
Nat Cell Biol ; 12(10): 982-92, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20818389

ABSTRACT

The epithelial-mesenchymal transition (EMT), one of the main mechanisms underlying development of cancer metastasis, induces stem-like properties in epithelial cells. Bmi1 is a polycomb-group protein that maintains self-renewal, and is frequently overexpressed in human cancers. Here, we show the direct regulation of BMI1 by the EMT regulator, Twist1. Furthermore, Twist1 and Bmi1 were mutually essential to promote EMT and tumour-initiating capability. Twist1 and Bmi1 act cooperatively to repress expression of both E-cadherin and p16INK4a. In patients with head and neck cancers, increased levels of both Twist1 and Bmi1 correlated with downregulation of E-cadherin and p16INK4a, and was associated with the worst prognosis. These results suggest that Twist1-induced EMT and tumour-initiating capability in cancer cells occurs through chromatin remodelling, which leads to unfavourable clinical outcomes.


Subject(s)
Epithelial Cells/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Mesoderm/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Twist-Related Protein 1/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Chromatin Assembly and Disassembly , Down-Regulation , Epithelial Cells/pathology , Exons/genetics , Female , Genes, p16 , Head and Neck Neoplasms/genetics , Humans , Male , Mesoderm/pathology , Middle Aged , Neoplasm Metastasis/genetics , Nuclear Proteins/genetics , Polycomb Repressive Complex 1 , Prognosis , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Transcription, Genetic , Twist-Related Protein 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...