Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 300(1): 105580, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38141763

ABSTRACT

Cancer cells acquire malignant phenotypes through an epithelial-mesenchymal transition, which is induced by environmental factors or extracellular signaling molecules, including transforming growth factor-ß (TGF-ß). Among epithelial-mesenchymal transition-associated cell responses, cell morphological changes and cell motility are closely associated with remodeling of the actin stress fibers. Here, we examined the TGF-ß signaling pathways leading to these cell responses. Through knockdown experiments in A549 lung adenocarcinoma cells, we found that Smad3-mediated induction of Snail, but not that of Slug, is indispensable for morphological changes, stress fiber formation, and enhanced motility in cells stimulated with TGF-ß. Ectopic expression of Snail in SMAD3-knockout cells rescued the defect in morphological changes and stress fiber formation by TGF-ß, indicating that the role of Smad3 in these responses is to upregulate Snail expression. Mechanistically, Snail is required for TGF-ß-induced upregulation of Wnt5b, which in turn activates RhoA and subsequent stress fiber formation in cooperation with phosphoinositide 3-kinase. However, ectopic expression of Snail in SMAD3-knockout cells failed to rescue the defect in cell motility enhancement by TGF-ß, indicating that activation of the Smad3/Snail/Wnt5b axis is indispensable but not sufficient for enhancing cell motility; a Smad3-dependent but Snail-independent pathway to activate Rac1 is additionally required. Therefore, the Smad3-dependent pathway leading to enhanced cell motility has two branches: a Snail-dependent branch to activate RhoA and a Snail-independent branch to activate Rac1. Coordinated activation of these branches, together with activation of non-Smad signaling pathways, mediates enhanced cell motility induced by TGF-ß.


Subject(s)
Signal Transduction , Smad3 Protein , Snail Family Transcription Factors , Stress Fibers , Transforming Growth Factor beta , rho GTP-Binding Proteins , Humans , A549 Cells , Cell Movement , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Phosphatidylinositol 3-Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Smad3 Protein/deficiency , Smad3 Protein/genetics , Smad3 Protein/metabolism , Snail Family Transcription Factors/deficiency , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Stress Fibers/metabolism , Transforming Growth Factor beta/metabolism , Enzyme Activation , Actins/metabolism , Mesoderm/metabolism , Mesoderm/pathology
2.
JCO Precis Oncol ; 52021 06.
Article in English | MEDLINE | ID: mdl-34322653

ABSTRACT

PURPOSE: Androgen receptor (AR) signaling is important in prostate cancer progression, and therapies that target this pathway have been the mainstay of treatment for advanced disease for over 70 years. Tumors eventually progress despite castration through a number of well-characterized mechanisms; however, little is known about what determines the magnitude of response to short-term pathway inhibition. METHODS: We evaluated a novel combination of AR-targeting therapies (degarelix, abiraterone, and bicalutamide) and noted that the objective patient response to therapy was highly variable. To investigate what was driving treatment resistance in poorly responding patients, as a secondary outcome we comprehensively characterized pre- and post-treatment samples using both whole-genome and RNA sequencing. RESULTS: We find that resistance following short-term treatment differs molecularly from typical progressive castration-resistant disease, associated with transcriptional reprogramming, to a transitional epithelial-to-mesenchymal transition (EMT) phenotype rather than an upregulation of AR signaling. Unexpectedly, tolerance to therapy appears to be the default state, with treatment response correlating with the prevalence of tumor cells deficient for SNAI2, a key regulator of EMT reprogramming. CONCLUSION: We show that EMT characterizes acutely resistant prostate tumors and that deletion of SNAI2, a key transcriptional regulator of EMT, correlates with clinical response.


Subject(s)
Androgen Antagonists/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Epithelial-Mesenchymal Transition/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Snail Family Transcription Factors/genetics , Aged , Androgen Antagonists/adverse effects , Androstenes , Anilides , Antineoplastic Agents, Hormonal/adverse effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Nitriles , Oligopeptides , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Signal Transduction , Snail Family Transcription Factors/deficiency , Tosyl Compounds
3.
J Vis Exp ; (168)2021 02 28.
Article in English | MEDLINE | ID: mdl-33720141

ABSTRACT

Somitogenesis is a hallmark of vertebrate embryonic development. For years, researchers have been studying this process in a variety of organisms using a wide range of techniques encompassing ex vivo and in vitro approaches. However, most studies still rely on the analysis of two-dimensional (2D) imaging data, which limits proper evaluation of a developmental process like axial extension and somitogenesis involving highly dynamic interactions in a complex 3D space. Here we describe techniques that allow mouse live imaging acquisition, dataset processing, visualization and analysis in 3D and 4D to study the cells (e.g., neuromesodermal progenitors) involved in these developmental processes. We also provide a step-by-step protocol for optical projection tomography and whole-mount immunofluorescence microscopy in mouse embryos (from sample preparation to image acquisition) and show a pipeline that we developed to process and visualize 3D image data. We extend the use of some of these techniques and highlight specific features of different available software (e.g., Fiji/ImageJ, Drishti, Amira and Imaris) that can be used to improve our current understanding of axial extension and somite formation (e.g., 3D reconstructions). Altogether, the techniques here described emphasize the importance of 3D data visualization and analysis in developmental biology, and might help other researchers to better address 3D and 4D image data in the context of vertebrate axial extension and segmentation. Finally, the work also employs novel tools to facilitate teaching vertebrate embryonic development.


Subject(s)
Body Patterning , Imaging, Three-Dimensional/methods , Vertebrates/anatomy & histology , Vertebrates/embryology , Animals , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/diagnostic imaging , Embryonic Development , Fluorescent Antibody Technique , Mice, Knockout , Snail Family Transcription Factors/deficiency , Snail Family Transcription Factors/metabolism , Software , Time Factors , Tissue Fixation , Tomography, Optical
4.
Cell Rep ; 28(2): 394-407.e6, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31291576

ABSTRACT

DNA damage activates checkpoints that limit the replicative potential of stem cells, including differentiation. These checkpoints protect against cancer development but also promote tissue aging. Because mice lacking Slug/Snai2 exhibit limited stem cell activity, including luminobasal differentiation, and are protected from mammary cancer, we reasoned that Slug might regulate DNA damage checkpoints in mammary epithelial cells. Here, we show that Slug facilitates efficient execution of RPA32-mediated DNA damage response (DDR) signaling. Slug deficiency leads to delayed phosphorylation of ataxia telangiectasia mutated and Rad3-related protein (ATR) and its effectors RPA32 and CHK1. This leads to impaired RAD51 recruitment to DNA damage sites and persistence of unresolved DNA damage. In vivo, Slug/Snai2 loss leads to increased DNA damage and premature aging of mammary epithelium. Collectively, our work demonstrates that the mammary stem cell regulator Slug controls DDR checkpoints by dually inhibiting differentiation and facilitating DDR repair, and its loss causes unresolved DNA damage and accelerated aging.


Subject(s)
DNA Damage , DNA Repair , Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Snail Family Transcription Factors/deficiency , Animals , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Cellular Senescence/physiology , HEK293 Cells , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
5.
Artif Cells Nanomed Biotechnol ; 45(5): 969-974, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27322434

ABSTRACT

Snail-1 known as one of the important transcription factor is a mediator of survival and cell migration, and expression is raised in numerous cancer types. Snail-1 gene may show a role in recurrence of several cancers including bladder cancer by down-regulating E-cadherin, inducing an epithelial to mesenchymal transition (EMT) and its related microRNAs (miRNAs). The aim of this study was to investigate the effect of a specific Snail-1 siRNA on apoptosis and alter EMT related miRNAs of EJ-138 (bladder cancer) cells. The cells were transfected with siRNAs using transfection reagent. The cytotoxic effects of Snail-1 siRNA, on bladder cancer cells were determined using MTT assay. Relative Snail-1 mRNA levels were measured by QRT- PCR, respectively. Apoptosis was measured by TUNEL test based on labeling of DNA strand breaks. We also evaluated miR-29b, miR-21, and miR-203 expression by QRT-PCR to determine alteration in miRNAs expression involved in EMT. Snail-1 siRNA significantly reduced mRNA expression levels in 48 h after transfection at the concentration of 60 pmol in bladder cancer cells. We also showed that the silencing of Snail-1 led to the induction of apoptosis. miR-21 and miR-29b depression have been shown in Snail-1 suppressed group in EJ-138 cells in vitro. These results propose that Snail-1 might play an important role in the progression of bladder cancer, and be a potential therapeutic target for trigger apoptosis and suppression of EMT-related miRNAs in bladder cancer.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , MicroRNAs/genetics , RNA, Small Interfering/genetics , Snail Family Transcription Factors/genetics , Urinary Bladder Neoplasms/pathology , Cadherins/genetics , Cell Line, Tumor , Down-Regulation/genetics , Humans , Neoplasm Invasiveness , Snail Family Transcription Factors/deficiency
6.
PLoS One ; 11(10): e0164752, 2016.
Article in English | MEDLINE | ID: mdl-27760172

ABSTRACT

China accounts for almost half of the total number of liver cancer cases and deaths worldwide, and hepatocellular carcinoma (HCC) is the most primary liver cancer. Snail family transcriptional repressor 2 (SNAI2) is known as an epithelial to mesenchymal transition-inducing transcription factor that drives neoplastic epithelial cells into mesenchymal phenotype. However, the roles of endogenous SNAI2 remain controversial in different types of malignant tumors. Herein, we surprisingly identify that anchorage-independent growth, including the formation of tumor sphere and soft agar colony, is significantly increased when SNAI2 expression is inhibited by shRNAs in HCC cells. Suppression of SNAI2 suffices to up-regulate several cancer stem genes. Although unrelated to the metastatic ability, SNAI2 inhibition does increase the efflux of Hoechst 33342 and enhance multidrug resistance in vitro and in vivo. In agreement with this data, we demonstrate for the first time that decreasing SNAI2 level can transcriptionally upregulate several ATP binding cassette (ABC) transporter genes such as ABCB1. Moreover, ABC transporters' inhibitor verapamil can rescue the multidrug resistance induced by SNAI2 inhibition. Our results implicate that SNAI2 behaves as a tumor suppressor by inhibiting multidrug resistance via suppressing ABC transporter genes in HCC cells.


Subject(s)
Carcinoma, Hepatocellular/pathology , Drug Resistance, Multiple/genetics , Liver Neoplasms/pathology , Snail Family Transcription Factors/deficiency , Snail Family Transcription Factors/genetics , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Resistance, Multiple/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Silencing , Humans , Mice , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Small Interfering/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , Verapamil/pharmacology
7.
PLoS One ; 11(10): e0162636, 2016.
Article in English | MEDLINE | ID: mdl-27706205

ABSTRACT

Hypoxic injury to the heart results in cardiac fibrosis that leads to cardiac dysfunction and heart failure. SNAIL1 is a zinc finger transcription factor implicated in fibrosis following organ injury and cancer. To determine if the action of SNAIL1 contributed to cardiac fibrosis following hypoxic injury, we used an endogenous SNAIL1 bioluminescence reporter mice, and SNAIL1 knockout mouse models. Here we report that SNAIL1 expression is upregulated in the infarcted heart, especially in the myofibroblasts. Utilizing primary cardiac fibroblasts in ex vivo cultures we find that pro-fibrotic factors and collagen I increase SNAIL1 protein level. SNAIL1 is required in cardiac fibroblasts for the adoption of myofibroblast fate, collagen I expression and expression of fibrosis-related genes. Taken together this data suggests that SNAIL1 expression is induced in the cardiac fibroblasts after hypoxic injury and contributes to myofibroblast phenotype and a fibrotic scar formation. Resultant collagen deposition in the scar can maintain elevated SNAIL1 expression in the myofibroblasts and help propagate fibrosis.


Subject(s)
Myocardial Infarction/pathology , Snail Family Transcription Factors/genetics , Animals , Aorta, Thoracic/surgery , Cell Hypoxia , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Disease Models, Animal , Extracellular Matrix/metabolism , Fibrosis , Fluorescent Antibody Technique , Heart/diagnostic imaging , Mice , Mice, Knockout , Myocardial Infarction/metabolism , Myocardium/metabolism , Myofibroblasts/cytology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Platelet-Derived Growth Factor/pharmacology , RNA, Messenger/metabolism , Snail Family Transcription Factors/deficiency , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology
8.
Dev Biol ; 414(2): 142-8, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27141871

ABSTRACT

Epithelial stem cells undergo constant self-renewal and differentiation to maintain the homeostasis of epithelial tissues that undergo rapid turnover. Recent studies have shown that the epithelial-mesenchymal transition (EMT), which is primarily mediated by Snail via the suppression of E-cadherin, is able to generate cells with stem cell properties. However, the role of Snail in epithelial stem cells remains unclear. Here, we report that Snail directly controls proliferation of follicle stem cells (FSCs) in Drosophila females. Disruption of Snail expression in FSCs compromises their proliferation, but not their maintenance. Conversely, FSCs with excessive Snail expression display increased proliferation and lifespan, which is accompanied by a moderate decrease in the expression of E-cadherin (required for adhesion of FSCs to their niche) at the junction between their adjacent cells, indicating a conserved role of Snail in E-cadherin inhibition, which promote epithelial cell proliferation. Interestingly, a decrease in E-cadherin in snail-knock down FSCs does not restore the decreased proliferation of snail-knock down FSCs, suggesting that adhesion strength of FSCs to their niche is dispensable for Snail-mediated FSC division. Our results demonstrate that Snail controls epithelial stem cell division independently of its known role in the EMT, which contributes to induction of cancer stem cells.


Subject(s)
Cadherins/biosynthesis , Drosophila Proteins/biosynthesis , Ovarian Follicle/cytology , Ovary/cytology , Snail Family Transcription Factors/physiology , Stem Cells/cytology , Animals , Cadherins/genetics , Cell Adhesion , Cell Division , Cellular Senescence , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Epithelial Cells/cytology , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Luminescent Proteins/analysis , Mosaicism , Snail Family Transcription Factors/deficiency , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL
...