Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
JCI Insight ; 8(20)2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37870961

ABSTRACT

Despite being in the same pathway, mutations of KRAS and BRAF in colorectal carcinomas (CRCs) determine distinct progression courses. ZEB1 induces an epithelial-to-mesenchymal transition (EMT) and is associated with worse progression in most carcinomas. Using samples from patients with CRC, mouse models of KrasG12D and BrafV600E CRC, and a Zeb1-deficient mouse, we show that ZEB1 had opposite functions in KRAS- and BRAF-mutant CRCs. In KrasG12D CRCs, ZEB1 was correlated with a worse prognosis and a higher number of larger and undifferentiated (mesenchymal or EMT-like) tumors. Surprisingly, in BrafV600E CRC, ZEB1 was associated with better prognosis; fewer, smaller, and more differentiated (reduced EMT) primary tumors; and fewer metastases. ZEB1 was positively correlated in KRAS-mutant CRC cells and negatively in BRAF-mutant CRC cells with gene signatures for EMT, cell proliferation and survival, and ERK signaling. On a mechanistic level, ZEB1 knockdown in KRAS-mutant CRC cells increased apoptosis and reduced clonogenicity and anchorage-independent growth; the reverse occurred in BRAFV600E CRC cells. ZEB1 is associated with better prognosis and reduced EMT signature in patients harboring BRAF CRCs. These data suggest that ZEB1 can function as a tumor suppressor in BRAF-mutant CRCs, highlighting the importance of considering the KRAS/BRAF mutational background of CRCs in therapeutic strategies targeting ZEB1/EMT.


Subject(s)
Carcinoma , Colorectal Neoplasms , Proto-Oncogene Proteins B-raf , Zinc Finger E-box-Binding Homeobox 1 , Animals , Humans , Mice , Colorectal Neoplasms/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
2.
Cancer Res ; 80(14): 2983-2995, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32503808

ABSTRACT

Epithelial-to-mesenchymal transition (EMT)-inducing transcription factors (TF) are well known for their ability to induce mesenchymal states associated with increased migratory and invasive properties. Unexpectedly, nuclear expression of the EMT-TF ZEB2 in human primary melanoma has been shown to correlate with reduced invasion. We report here that ZEB2 is required for outgrowth for primary melanomas and metastases at secondary sites. Ablation of Zeb2 hampered outgrowth of primary melanomas in vivo, whereas ectopic expression enhanced proliferation and growth at both primary and secondary sites. Gain of Zeb2 expression in pulmonary-residing melanoma cells promoted the development of macroscopic lesions. In vivo fate mapping made clear that melanoma cells undergo a conversion in state where ZEB2 expression is replaced by ZEB1 expression associated with gain of an invasive phenotype. These findings suggest that reversible switching of the ZEB2/ZEB1 ratio enhances melanoma metastatic dissemination. SIGNIFICANCE: ZEB2 function exerts opposing behaviors in melanoma by promoting proliferation and expansion and conversely inhibiting invasiveness, which could be of future clinical relevance. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/14/2983/F1.large.jpg.


Subject(s)
Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Lung Neoplasms/secondary , Melanoma/pathology , Transcription Factors/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Neoplasm Invasiveness , Transcription Factors/genetics , Tumor Cells, Cultured , Zinc Finger E-box Binding Homeobox 2/genetics
3.
Gut ; 68(12): 2129-2141, 2019 12.
Article in English | MEDLINE | ID: mdl-31366457

ABSTRACT

OBJECTIVE: Chronic inflammation is a risk factor in colorectal cancer (CRC) and reactive oxygen species (ROS) released by the inflamed stroma elicit DNA damage in epithelial cells. We sought to identify new drivers of ulcerative colitis (UC) and inflammatory CRC. DESIGN: The study uses samples from patients with UC, mouse models of colitis and CRC and mice deficient for the epithelial-to-mesenchymal transition factor ZEB1 and the DNA repair glycosylase N-methyl-purine glycosylase (MPG). Samples were analysed by immunostaining, qRT-PCR, chromatin immunoprecipitation assays, microbiota next-generation sequencing and ROS determination. RESULTS: ZEB1 was induced in the colonic epithelium of UC and of mouse models of colitis. Compared with wild-type counterparts, Zeb1-deficient mice were partially protected from experimental colitis and, in a model of inflammatory CRC, they developed fewer tumours and exhibited lower levels of DNA damage (8-oxo-dG) and higher expression of MPG. Knockdown of ZEB1 in CRC cells inhibited 8-oxo-dG induction by oxidative stress (H2O2) and inflammatory cytokines (interleukin (IL)1ß). ZEB1 bound directly to the MPG promoter whose expression inhibited. This molecular mechanism was validated at the genetic level and the crossing of Zeb1-deficient and Mpg-deficient mice reverted the reduced inflammation and tumourigenesis in the former. ZEB1 expression in CRC cells induced ROS and IL1ß production by macrophages that, in turn, lowered MPG in CRC cells thus amplifying a positive loop between both cells to promote DNA damage and inhibit DNA repair. CONCLUSIONS: ZEB1 promotes colitis and inflammatory CRC through the inhibition of MPG in epithelial cells, thus offering new therapeutic strategies to modulate inflammation and inflammatory cancer.


Subject(s)
Colitis, Ulcerative/genetics , Colonic Neoplasms/genetics , DNA Glycosylases/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms, Experimental , Zinc Finger E-box-Binding Homeobox 1/genetics , Animals , Biopsy , Cells, Cultured , Colitis, Ulcerative/complications , Colitis, Ulcerative/metabolism , Colonic Neoplasms/etiology , Colonic Neoplasms/pathology , DNA Glycosylases/metabolism , DNA Repair , Epithelial Cells/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Neoplasm/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Fingers
4.
Nat Commun ; 10(1): 1364, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30910999

ABSTRACT

The mechanisms linking muscle injury and regeneration are not fully understood. Here we report an unexpected role for ZEB1 regulating inflammatory and repair responses in dystrophic and acutely injured muscles. ZEB1 is upregulated in the undamaged and regenerating myofibers of injured muscles. Compared to wild-type counterparts, Zeb1-deficient injured muscles exhibit enhanced damage that corresponds with a retarded p38-MAPK-dependent transition of their macrophages towards an anti-inflammatory phenotype. Zeb1-deficient injured muscles also display a delayed and poorer regeneration that is accounted by the retarded anti-inflammatory macrophage transition and their intrinsically deficient muscle satellite cells (MuSCs). Macrophages in Zeb1-deficient injured muscles show lower phosphorylation of p38 and its forced activation reverts the enhanced muscle damage and poorer regeneration. MuSCs require ZEB1 to maintain their quiescence, prevent their premature activation following injury, and drive efficient regeneration in dystrophic muscles. These data indicate that ZEB1 protects muscle from damage and is required for its regeneration.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , RNA, Messenger/genetics , Regeneration/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chromones/pharmacology , Disease Models, Animal , Flavonoids/pharmacology , Gene Expression Regulation , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/immunology , Laminin/genetics , Laminin/immunology , Macrophages/immunology , Macrophages/pathology , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Morpholines/pharmacology , Muscle, Skeletal/immunology , Muscle, Skeletal/injuries , Muscular Dystrophies/immunology , Muscular Dystrophies/pathology , Phenotype , Phosphorylation , RNA, Messenger/immunology , Regeneration/immunology , Satellite Cells, Skeletal Muscle/immunology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Signal Transduction , Zinc Finger E-box-Binding Homeobox 1/deficiency , Zinc Finger E-box-Binding Homeobox 1/immunology , p38 Mitogen-Activated Protein Kinases/immunology
5.
Nucleic Acids Res ; 46(20): 10697-10708, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30304480

ABSTRACT

Multiple physiopathological and clinical conditions trigger skeletal muscle atrophy through the induction of a group of proteins (atrogenes) that includes components of the ubiquitin-proteasome and autophagy-lysosomal systems. Atrogenes are induced by FOXO transcription factors, but their regulation is still not fully understood. Here, we showed that the transcription factor ZEB1, best known for promoting tumor progression, inhibits muscle atrophy and atrogene expression by antagonizing FOXO3-mediated induction of atrogenes. Compared to wild-type counterparts, hindlimb immobilization in Zeb1-deficient mice resulted in enhanced muscle atrophy and higher expression of a number of atrogenes, including Atrogin-1/Fbxo32, MuRF1/Trim63, Ctsl, 4ebp1, Gabarapl1, Psma1 and Nrf2. Likewise, in the C2C12 myogenic cell model, ZEB1 knockdown augmented both myotube diameter reduction and atrogene upregulation in response to nutrient deprivation. Mechanistically, ZEB1 directly represses in vitro and in vivo Fbxo32 and Trim63 promoter transcription in a stage-dependent manner and in a reverse pattern with MYOD1. ZEB1 bound to the Fbxo32 promoter in undifferentiated myoblasts and atrophic myotubes, but not in non-atrophic myotubes, where it is displaced by MYOD1. ZEB1 repressed both promoters through CtBP-mediated inhibition of FOXO3 transcriptional activity. These results set ZEB1 as a new target in therapeutic approaches to clinical conditions causing muscle mass loss.


Subject(s)
Alcohol Oxidoreductases/genetics , DNA-Binding Proteins/genetics , Forkhead Box Protein O3/genetics , Gene Expression Regulation , Muscular Atrophy/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Alcohol Oxidoreductases/metabolism , Animals , Cell Differentiation , Cell Line , DNA-Binding Proteins/metabolism , Forkhead Box Protein O3/metabolism , HEK293 Cells , Homeostasis , Humans , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Myoblasts/metabolism , Neoplasms/metabolism , Promoter Regions, Genetic , SKP Cullin F-Box Protein Ligases/metabolism , Transcription, Genetic , Zinc Finger E-box-Binding Homeobox 1/metabolism
6.
Nat Commun ; 8(1): 1758, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29170450

ABSTRACT

Notch1 transactivates Notch3 to drive terminal differentiation in stratified squamous epithelia. Notch1 and other Notch receptor paralogs cooperate to act as a tumor suppressor in squamous cell carcinomas (SCCs). However, Notch1 can be stochastically activated to promote carcinogenesis in murine models of SCC. Activated form of Notch1 promotes xenograft tumor growth when expressed ectopically. Here, we demonstrate that Notch1 activation and epithelial-mesenchymal transition (EMT) are coupled to promote SCC tumor initiation in concert with transforming growth factor (TGF)-ß present in the tumor microenvironment. We find that TGFß activates the transcription factor ZEB1 to repress Notch3, thereby limiting terminal differentiation. Concurrently, TGFß drives Notch1-mediated EMT to generate tumor initiating cells characterized by high CD44 expression. Moreover, Notch1 is activated in a small subset of SCC cells at the invasive tumor front and predicts for poor prognosis of esophageal SCC, shedding light upon the tumor promoting oncogenic aspect of Notch1 in SCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Epithelial-Mesenchymal Transition , Esophageal Squamous Cell Carcinoma/metabolism , Receptor, Notch1/metabolism , Receptor, Notch3/metabolism , Animals , Carcinogenesis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/physiopathology , Cell Line, Tumor , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/physiopathology , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Mice, Nude , Mice, Transgenic , Receptor, Notch1/genetics , Receptor, Notch3/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
7.
EMBO J ; 36(22): 3336-3355, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29038174

ABSTRACT

Accumulation of tumor-associated macrophages (TAMs) associates with malignant progression in cancer. However, the mechanisms that drive the pro-tumor functions of TAMs are not fully understood. ZEB1 is best known for driving an epithelial-to-mesenchymal transition (EMT) in cancer cells to promote tumor progression. However, a role for ZEB1 in macrophages and TAMs has not been studied. Here we describe that TAMs require ZEB1 for their tumor-promoting and chemotherapy resistance functions in a mouse model of ovarian cancer. Only TAMs that expressed full levels of Zeb1 accelerated tumor growth. Mechanistically, ZEB1 expression in TAMs induced their polarization toward an F4/80low pro-tumor phenotype, including direct activation of Ccr2 In turn, expression of ZEB1 by TAMs induced Ccl2, Cd74, and a mesenchymal/stem-like phenotype in cancer cells. In human ovarian carcinomas, TAM infiltration and CCR2 expression correlated with ZEB1 in tumor cells, where along with CCL2 and CD74 determined poorer prognosis. Importantly, ZEB1 in TAMs was a factor of poorer survival in human ovarian carcinomas. These data establish ZEB1 as a key factor in the tumor microenvironment and for maintaining TAMs' tumor-promoting functions.


Subject(s)
Carcinogenesis/pathology , Macrophages/metabolism , Macrophages/pathology , Neoplasms/metabolism , Neoplasms/pathology , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Count , Cell Differentiation/drug effects , Chemokine CCL2/pharmacology , Colony-Stimulating Factors/pharmacology , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class II/metabolism , Humans , Macrophage Activation/drug effects , Macrophages/drug effects , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Models, Biological , Monocytes/drug effects , Monocytes/pathology , Neoplasms/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phenotype , Receptors, CCR2/metabolism , Survival Analysis , Up-Regulation/drug effects
8.
Sci Rep ; 7(1): 45, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28246385

ABSTRACT

Human uveal melanoma (UM) is a major ocular malignant tumor with high risk of metastasis and requires multiple oncogenic factors for progression. ZEB1 is a zinc finger E-box binding transcription factor known for participating epithelial-mesenchymal transition (EMT), a critical cellular event for metastasis of malignant tumors of epithelium origin. ZEB1 is also expressed in UM and high expression of ZEB1 correlates with UM advancement, but has little effect on cell morphology. We show that spindle UM cells can become epithelioid but not vice versa; and ZEB1 exerts its tumorigenic effects by promoting cell dedifferentiation, proliferation, invasiveness, and dissemination. We provide evidence that ZEB1 binds not only to repress critical genes involving in pigment synthesis, mitosis, adherent junctions, but also to transactivate genes involving in matrix degradation and cellular locomotion to propel UM progression towards metastasis. We conclude that ZEB1 is a major oncogenic factor required for UM progression and could be a potential therapeutic target for treating UM in the clinic.


Subject(s)
Melanoma/genetics , Melanoma/metabolism , Uveal Neoplasms/genetics , Uveal Neoplasms/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Animals , Carcinogenesis , Cell Dedifferentiation , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Melanoma/pathology , Mice, Nude , Neoplasm Invasiveness , Oncogenes , Uveal Neoplasms/pathology , Zinc Finger E-box-Binding Homeobox 1/metabolism
9.
Gut ; 66(4): 666-682, 2017 04.
Article in English | MEDLINE | ID: mdl-27965283

ABSTRACT

OBJECTIVE: Understand the role of ZEB1 in the tumour initiation and progression beyond inducing an epithelial-to-mesenchymal transition. DESIGN: Expression of the transcription factor ZEB1 associates with a worse prognosis in most cancers, including colorectal carcinomas (CRCs). The study uses survival analysis, in vivo mouse transgenic and xenograft models, gene expression arrays, immunostaining and gene and protein regulation assays. RESULTS: The poorer survival determined by ZEB1 in CRCs depended on simultaneous high levels of the Wnt antagonist DKK1, whose expression was transcriptionally activated by ZEB1. In cancer cells with mutant TP53, ZEB1 blocked the formation of senescence-associated heterochromatin foci at the onset of senescence by triggering a new regulatory cascade that involves the subsequent activation of DKK1, mutant p53, Mdm2 and CtBP to ultimately repress macroH2A1 (H2AFY). In a transgenic mouse model of colon cancer, partial downregulation of Zeb1 was sufficient to induce H2afy and to trigger in vivo tumour senescence, thus resulting in reduced tumour load and improved survival. The capacity of ZEB1 to induce tumourigenesis in a xenograft mouse model requires the repression of H2AFY by ZEB1. Lastly, the worst survival effect of ZEB1 in patients with CRC ultimately depends on low expression of H2AFY and of senescence-associated genes. CONCLUSIONS: The tumourigenic capacity of ZEB1 depends on its inhibition of cancer cell senescence through the activation of a herein identified new molecular pathway. These results set ZEB1 as a potential target in therapeutic strategies aimed at inducing senescence.


Subject(s)
Carcinogenesis/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Histones/genetics , Intercellular Signaling Peptides and Proteins/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Cell Line, Tumor , Cell Survival/genetics , Cellular Senescence/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Heterografts , Histones/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Survival Rate , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Wnt Signaling Pathway , Zinc Finger E-box-Binding Homeobox 1/metabolism
10.
Cell Rep ; 17(6): 1473-1481, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27806288

ABSTRACT

During spinal cord development, astrocyte precursors arise from neuroepithelial progenitors, delaminate from the ventricular zone, and migrate toward their final locations where they differentiate. Although the mechanisms underlying their early specification and late differentiation are being deciphered, less is known about the temporal control of their migration. Here, we show that the epithelial-mesenchymal transition regulator Zeb1 is expressed in glial precursors and report that loss of Zeb1 function specifically delays the onset of astrocyte precursor delamination from the ventricular zone, correlating with transient deregulation of the adhesion protein Cadherin-1. Consequently, astrocyte precursor invasion into the Zeb1-/- mutant white matter is delayed, and induction of their differentiation is postponed. These findings illustrate how fine regulation of adhesive properties influences the onset of neural precursor migration and further support the notion that duration of exposure of migrating astrocyte precursors to environmental cues and/or their correct positioning influence the timing of their differentiation.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Cell Movement , Spinal Cord/cytology , Stem Cells/cytology , Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Aging/genetics , Animals , Body Patterning , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice , Mutation/genetics
11.
J Cell Physiol ; 231(10): 2205-17, 2016 10.
Article in English | MEDLINE | ID: mdl-26868487

ABSTRACT

ZEB1 transcription factor is important in both development and disease, including many TGFß-induced responses, and the epithelial-to-mesenchymal transition (EMT) by which many tumors undergo metastasis. ZEB1 is differentially phosphorylated in different cell types; however the role of phosphorylation in ZEB1 activity is unknown. Luciferase reporter studies and electrophoresis mobility shift assays (EMSA) show that a decrease in phosphorylation of ZEB1 increases both DNA-binding and transcriptional repression of ZEB1 target genes. Functional analysis of ZEB1 phosphorylation site mutants near the second zinc finger domain (termed ZD2) show that increased phosphorylation (due to either PMA plus ionomycin, or IGF-1) can inhibit transcriptional repression by either a ZEB1-ZD2 domain clone, or full-length ZEB1. This approach identifies phosphosites that have a substantial effect regulating the transcriptional and DNA-binding activity of ZEB1. Immunoprecipitation with anti-ZEB1 antibodies followed by western analysis with a phospho-Threonine-Proline-specific antibody indicates that the ERK consensus site at Thr-867 is phosphorylated in ZEB1. In addition to disrupting in vitro DNA-binding measured by EMSA, IGF-1-induced MEK/ERK phosphorylation is sufficient to disrupt nuclear localization of GFP-ZEB1 fusion clones. These data suggest that phosphorylation of ZEB1 integrates TGFß signaling with other signaling pathways such as IGF-1. J. Cell. Physiol. 231: 2205-2217, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , CHO Cells , Cricetulus , Epithelial-Mesenchymal Transition , Insulin-Like Growth Factor I/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Signal Transduction/physiology , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Fingers
12.
Cell Microbiol ; 18(6): 844-58, 2016 06.
Article in English | MEDLINE | ID: mdl-26639759

ABSTRACT

The oral anaerobe Porphyromonas gingivalis is associated with the development of cancers including oral squamous cell carcinoma (OSCC). Here, we show that infection of gingival epithelial cells with P. gingivalis induces expression and nuclear localization of the ZEB1 transcription factor, which controls epithelial-mesenchymal transition. P. gingivalis also caused an increase in ZEB1 expression as a dual species community with Fusobacterium nucleatum or Streptococcus gordonii. Increased ZEB1 expression was associated with elevated ZEB1 promoter activity and did not require suppression of the miR-200 family of microRNAs. P. gingivalis strains lacking the FimA fimbrial protein were attenuated in their ability to induce ZEB1 expression. ZEB1 levels correlated with an increase in expression of mesenchymal markers, including vimentin and MMP-9, and with enhanced migration of epithelial cells into matrigel. Knockdown of ZEB1 with siRNA prevented the P. gingivalis-induced increase in mesenchymal markers and epithelial cell migration. Oral infection of mice by P. gingivalis increased ZEB1 levels in gingival tissues, and intracellular P. gingivalis were detected by antibody staining in biopsy samples from OSCC. These findings indicate that FimA-driven ZEB1 expression could provide a mechanistic basis for a P. gingivalis contribution to OSCC.


Subject(s)
Gingiva/microbiology , Porphyromonas gingivalis/pathogenicity , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Cell Movement , Epithelial Cells/microbiology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Fimbriae, Bacterial/metabolism , Gene Expression Regulation , Gingiva/cytology , Gingiva/metabolism , Host-Pathogen Interactions , Humans , Keratinocytes/microbiology , Keratinocytes/pathology , Mice, Inbred BALB C , MicroRNAs/genetics , Mouth Neoplasms/microbiology , Porphyromonas gingivalis/genetics , Promoter Regions, Genetic , Zinc Finger E-box-Binding Homeobox 1/genetics
13.
Genom Data ; 5: 82-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26484231

ABSTRACT

In order to understand the process of terminal differentiation in salivary acinar cells, mRNA and microRNA expression was measured across the month long process of differentiation in the parotid gland of the rat. Acinar cells were isolated at either nine time points (mRNA) or four time points (microRNA) in triplicate using laser capture microdissection (LCM). One of the values of this dataset comes from the high quality RNA (RIN > 7) that was used in this study, which can be prohibitively difficult to obtain from such an RNaseI-rich tissue. Global mRNA expression was measured by rat genome microarray hybridization (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE65586), and expression of microRNAs by qPCR array (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE65324). Comparing expression at different ages, 2656 mRNAs and 64 microRNAs were identified as differentially expressed. Because mRNA expression was sampled at many time points, clustering and regression analysis were able to identify dynamic expression patterns that had not been implicated in acinar differentiation before. Integration of the two datasets allowed the identification of microRNA target genes, and a gene regulatory network. Bioinformatics R code and additional details of experimental methods and data analysis are provided.

14.
PLoS One ; 10(4): e0125153, 2015.
Article in English | MEDLINE | ID: mdl-25928148

ABSTRACT

OBJECTIVE: The transcription factor networks that drive parotid salivary gland progenitor cells to terminally differentiate, remain largely unknown and are vital to understanding the regeneration process. METHODOLOGY: A systems biology approach was taken to measure mRNA and microRNA expression in vivo across acinar cell terminal differentiation in the rat parotid salivary gland. Laser capture microdissection (LCM) was used to specifically isolate acinar cell RNA at times spanning the month-long period of parotid differentiation. RESULTS: Clustering of microarray measurements suggests that expression occurs in four stages. mRNA expression patterns suggest a novel role for Pparg which is transiently increased during mid postnatal differentiation in concert with several target gene mRNAs. 79 microRNAs are significantly differentially expressed across time. Profiles of statistically significant changes of mRNA expression, combined with reciprocal correlations of microRNAs and their target mRNAs, suggest a putative network involving Klf4, a differentiation inhibiting transcription factor, which decreases as several targeting microRNAs increase late in differentiation. The network suggests a molecular switch (involving Prdm1, Sox11, Pax5, miR-200a, and miR-30a) progressively decreases repression of Xbp1 gene transcription, in concert with decreased translational repression by miR-214. The transcription factor Xbp1 mRNA is initially low, increases progressively, and may be maintained by a positive feedback loop with Atf6. Transfection studies show that Xbp1 activates the Mist1 promoter [corrected]. In addition, Xbp1 and Mist1 each activate the parotid secretory protein (Psp) gene, which encodes an abundant salivary protein, and is a marker of terminal differentiation. CONCLUSION: This study identifies novel expression patterns of Pparg, Klf4, and Sox11 during parotid acinar cell differentiation, as well as numerous differentially expressed microRNAs. Network analysis identifies a novel stemness arm, a genetic switch involving transcription factors and microRNAs, and transition to an Xbp1 driven differentiation network. This proposed network suggests key regulatory interactions in parotid gland terminal differentiation.


Subject(s)
Acinar Cells/cytology , Parotid Gland/cytology , Systems Biology/methods , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , DNA-Binding Proteins/genetics , Female , Gene Regulatory Networks/genetics , Kruppel-Like Factor 4 , Pregnancy , RNA, Messenger/genetics , Rats , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , X-Box Binding Protein 1
16.
Nat Commun ; 5: 5660, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25434817

ABSTRACT

Ras pathway mutation is frequent in carcinomas where it induces expression of the transcriptional repressor ZEB1. Although ZEB1 is classically linked to epithelial-mesenchymal transition and tumour metastasis, it has an emerging second role in generation of cancer-initiating cells. Here we show that Ras induction of ZEB1 is required for tumour initiation in a lung cancer model, and we link this function to repression Pten, whose loss is critical for emergence of cancer-initiating cells. These two roles for ZEB1 in tumour progression can be distinguished by their requirement for different levels of ZEB1. A lower threshold of ZEB1 is sufficient for cancer initiation, whereas further induction is necessary for tumour metastasis.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Lung Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/metabolism , Adenocarcinoma/pathology , Adenoma/pathology , Animals , Disease Models, Animal , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Zinc Finger E-box-Binding Homeobox 1
17.
J Comput Biol ; 20(7): 524-39, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23829652

ABSTRACT

In modern systems biology the modeling of longitudinal data, such as changes in mRNA concentrations, is often of interest. Fully parametric, ordinary differential equations (ODE)-based models are typically developed for the purpose, but their lack of fit in some examples indicates that more flexible Bayesian models may be beneficial, particularly when there are relatively few data points available. However, under such sparse data scenarios it is often difficult to identify the most suitable model. The process of falsifying inappropriate candidate models is called model discrimination. We propose here a formal method of discrimination between competing Bayesian mixture-type longitudinal models that is both sensitive and sufficiently flexible to account for the complex variability of the longitudinal molecular data. The ideas from the field of Bayesian analysis of computer model validation are applied, along with modern Markov Chain Monte Carlo (MCMC) algorithms, in order to derive an appropriate Bayes discriminant rule. We restrict attention to the two-model comparison problem and present the application of the proposed rule to the mRNA data in the de-differentiation network of three mRNA concentrations in mammalian salivary glands as well as to a large synthetic dataset derived from the model used in the recent DREAM6 competition.


Subject(s)
Algorithms , Amylases/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Models, Statistical , Parotid Gland/cytology , RNA, Messenger/genetics , Salivary Proteins and Peptides/genetics , Amylases/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bayes Theorem , Humans , Markov Chains , Molecular Dynamics Simulation , Monte Carlo Method , Parotid Gland/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Salivary Proteins and Peptides/metabolism , Time Factors
18.
Mol Cell Biol ; 33(7): 1368-82, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23339872

ABSTRACT

Skeletal muscle development is orchestrated by the myogenic regulatory factor MyoD, whose activity is blocked in myoblasts by proteins preventing its nuclear translocation and/or binding to G/C-centered E-boxes in target genes. Recent evidence indicates that muscle gene expression is also regulated at the cis level by differential affinity for DNA between MyoD and other E-box binding proteins during myogenesis. MyoD binds to G/C-centered E-boxes, enriched in muscle differentiation genes, in myotubes but not in myoblasts. Here, we used cell-based and in vivo Drosophila, Xenopus laevis, and mouse models to show that ZEB1, a G/C-centered E-box binding transcriptional repressor, imposes a temporary stage-dependent inhibition of muscle gene expression and differentiation via CtBP-mediated transcriptional repression. We found that, contrary to MyoD, ZEB1 binds to G/C-centered E-boxes in muscle differentiation genes at the myoblast stage but not in myotubes. Its knockdown results in precocious expression of muscle differentiation genes and acceleration of myotube formation. Inhibition of muscle genes by ZEB1 occurs via transcriptional repression and involves recruitment of the CtBP corepressor. Lastly, we show that the pattern of gene expression associated with muscle differentiation is accelerated in ZEB1(-/-) mouse embryos. These results set ZEB1 as an important regulator of the temporal pattern of gene expression controlling muscle differentiation.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation , Homeodomain Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Muscle, Skeletal/physiology , Transcriptional Activation , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E-Box Elements , Gene Expression , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Xenopus laevis , Zinc Finger E-box-Binding Homeobox 1
19.
Clin Cancer Res ; 19(5): 1071-82, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23340304

ABSTRACT

PURPOSE: Carcinoma cells enhance their invasive capacity through dedifferentiation and dissolution of intercellular adhesions. A key activator of this process is the ZEB1 transcription factor, which is induced in invading cancer cells by canonical Wnt signaling (ß-catenin/TCF4). Tumor invasiveness also entails proteolytic remodeling of the peritumoral stroma. This study aimed to investigate the potential regulation by ZEB1 of the plasminogen proteolytic system constituted by the urokinase plasminogen activator (uPA), and its inhibitor, plasminogen activator inhibitor-1 (PAI-1). EXPERIMENTAL DESIGN: Through multiple experimental approaches, colorectal carcinoma (CRC) cell lines and samples from human primary CRC and ZEB1 (-/-) mice were used to examine ZEB1-mediated regulation of uPA and PAI-1 at the protein, mRNA, and transcriptional level. RESULTS: ZEB1 regulates uPA and PAI-1 in opposite directions: induces uPA and inhibits PAI-1. In vivo expression of uPA depends on ZEB1 as it is severely reduced in the developing intestine of ZEB1 null (-/-) mice. Optimal induction of uPA by Wnt signaling requires ZEB1 expression. ZEB1 binds to the uPA promoter and activates its transcription through a mechanism implicating the histone acetyltransferase p300. In contrast, inhibition of PAI-1 by ZEB1 does not involve transcriptional repression but rather downregulation of mRNA stability. ZEB1-mediated tumor cell migration and invasion depend on its induction of uPA. ZEB1 coexpresses with uPA in cancer cells at the invasive front of CRCs. CONCLUSIONS: ZEB1 promotes tumor invasiveness not only via induction in cancer cells of a motile dedifferentiated phenotype but also by differential regulation of genes involved in stroma remodeling.


Subject(s)
Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/physiology , Kruppel-Like Transcription Factors/physiology , Plasminogen Activator Inhibitor 1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Blotting, Western , Cell Movement , Cells, Cultured , Chromatin Immunoprecipitation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Plasminogen Activator Inhibitor 1/genetics , Promoter Regions, Genetic , RNA Stability , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Urokinase-Type Plasminogen Activator/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , Zinc Finger E-box-Binding Homeobox 1 , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
20.
Cell Mol Life Sci ; 69(20): 3429-56, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22945800

ABSTRACT

Cancer is a complex multistep process involving genetic and epigenetic changes that eventually result in the activation of oncogenic pathways and/or inactivation of tumor suppressor signals. During cancer progression, cancer cells acquire a number of hallmarks that promote tumor growth and invasion. A crucial mechanism by which carcinoma cells enhance their invasive capacity is the dissolution of intercellular adhesions and the acquisition of a more motile mesenchymal phenotype as part of an epithelial-to-mesenchymal transition (EMT). Although many transcription factors can trigger it, the full molecular reprogramming occurring during an EMT is mainly orchestrated by three major groups of transcription factors: the ZEB, Snail and Twist families. Upregulated expression of these EMT-activating transcription factors (EMT-ATFs) promotes tumor invasiveness in cell lines and xenograft mice models and has been associated with poor clinical prognosis in human cancers. Evidence accumulated in the last few years indicates that EMT-ATFs also regulate an expanding set of cancer cell capabilities beyond tumor invasion. Thus, EMT-ATFs have been shown to cooperate in oncogenic transformation, regulate cancer cell stemness, override safeguard programs against cancer like apoptosis and senescence, determine resistance to chemotherapy and promote tumor angiogenesis. This article reviews the expanding portfolio of functions played by EMT-ATFs in cancer progression.


Subject(s)
Activating Transcription Factors/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition , Neoplasms/metabolism , Neoplasms/pathology , Animals , Humans , Mice , Neoplasm Invasiveness
SELECTION OF CITATIONS
SEARCH DETAIL
...