Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 128(11): 4924-4937, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30130254

ABSTRACT

Mutant KRAS drives glycolytic flux in lung cancer, potentially impacting aberrant protein glycosylation. Recent evidence suggests aberrant KRAS drives flux of glucose into the hexosamine biosynthetic pathway (HBP). HBP is required for various glycosylation processes, such as protein N- or O-glycosylation and glycolipid synthesis. However, its function during tumorigenesis is poorly understood. One contributor and proposed target of KRAS-driven cancers is a developmentally conserved epithelial plasticity program called epithelial-mesenchymal transition (EMT). Here we showed in novel autochthonous mouse models that EMT accelerated KrasG12D lung tumorigenesis by upregulating expression of key enzymes of the HBP pathway. We demonstrated that HBP was required for suppressing KrasG12D-induced senescence, and targeting HBP significantly delayed KrasG12D lung tumorigenesis. To explore the mechanism, we investigated protein glycosylation downstream of HBP and found elevated levels of O-linked ß-N-acetylglucosamine (O-GlcNAcylation) posttranslational modification on intracellular proteins. O-GlcNAcylation suppressed KrasG12D oncogene-induced senescence (OIS) and accelerated lung tumorigenesis. Conversely, loss of O-GlcNAcylation delayed lung tumorigenesis. O-GlcNAcylation of proteins SNAI1 and c-MYC correlated with the EMT-HBP axis and accelerated lung tumorigenesis. Our results demonstrated that O-GlcNAcylation was sufficient and required to accelerate KrasG12D lung tumorigenesis in vivo, which was reinforced by epithelial plasticity programs.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/enzymology , Mutation, Missense , Protein Processing, Post-Translational , Proto-Oncogene Proteins p21(ras)/metabolism , A549 Cells , Acylation , Amino Acid Substitution , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Glucose/genetics , Glucose/metabolism , HEK293 Cells , Hexosamines/genetics , Hexosamines/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Mice, Transgenic , Proto-Oncogene Proteins p21(ras)/genetics
2.
Cancer Res ; 77(12): 3181-3193, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28484075

ABSTRACT

TWIST1 is a transcription factor critical for development that can promote prostate cancer metastasis. During embryonic development, TWIST1 and HOXA9 are coexpressed in mouse prostate and then silenced postnatally. Here we report that TWIST1 and HOXA9 coexpression are reactivated in mouse and human primary prostate tumors and are further enriched in human metastases, correlating with survival. TWIST1 formed a complex with WDR5 and the lncRNA Hottip/HOTTIP, members of the MLL/COMPASS-like H3K4 methylases, which regulate chromatin in the Hox/HOX cluster during development. TWIST1 overexpression led to coenrichment of TWIST1 and WDR5 as well as increased H3K4me3 chromatin at the Hoxa9/HOXA9 promoter, which was dependent on WDR5. Expression of WDR5 and Hottip/HOTTIP was also required for TWIST1-induced upregulation of HOXA9 and aggressive cellular phenotypes such as invasion and migration. Pharmacologic inhibition of HOXA9 prevented TWIST1-induced aggressive prostate cancer cellular phenotypes in vitro and metastasis in vivo This study demonstrates a novel mechanism by which TWIST1 regulates chromatin and gene expression by cooperating with the COMPASS-like complex to increase H3K4 trimethylation at target gene promoters. Our findings highlight a TWIST1-HOXA9 embryonic prostate developmental program that is reactivated during prostate cancer metastasis and is therapeutically targetable. Cancer Res; 77(12); 3181-93. ©2017 AACR.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/metabolism , Neoplasm Invasiveness/genetics , Nuclear Proteins/metabolism , Prostatic Neoplasms/pathology , RNA, Long Noncoding/metabolism , Twist-Related Protein 1/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Chromatin , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/genetics , Gene Knockout Techniques , Heterografts , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Male , Mice , Neoplasm Invasiveness/pathology , Nuclear Proteins/genetics , Polymerase Chain Reaction , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Twist-Related Protein 1/genetics
3.
Neoplasia ; 17(1): 16-31, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25622896

ABSTRACT

The TWIST1 gene has diverse roles in development and pathologic diseases such as cancer. TWIST1 is a dimeric basic helix-loop-helix (bHLH) transcription factor existing as TWIST1-TWIST1 or TWIST1-E12/47. TWIST1 partner choice and DNA binding can be influenced during development by phosphorylation of Thr125 and Ser127 of the Thr-Gln-Ser (TQS) motif within the bHLH of TWIST1. The significance of these TWIST1 phosphorylation sites for metastasis is unknown. We created stable isogenic prostate cancer cell lines overexpressing TWIST1 wild-type, phospho-mutants, and tethered versions. We assessed these isogenic lines using assays that mimic stages of cancer metastasis. In vitro assays suggested the phospho-mimetic Twist1-DQD mutation could confer cellular properties associated with pro-metastatic behavior. The hypo-phosphorylation mimic Twist1-AQA mutation displayed reduced pro-metastatic activity compared to wild-type TWIST1 in vitro, suggesting that phosphorylation of the TWIST1 TQS motif was necessary for pro-metastatic functions. In vivo analysis demonstrates that the Twist1-AQA mutation exhibits reduced capacity to contribute to metastasis, whereas the expression of the Twist1-DQD mutation exhibits proficient metastatic potential. Tethered TWIST1-E12 heterodimers phenocopied the Twist1-DQD mutation for many in vitro assays, suggesting that TWIST1 phosphorylation may result in heterodimerization in prostate cancer cells. Lastly, the dual phosphatidylinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) inhibitor BEZ235 strongly attenuated TWIST1-induced migration that was dependent on the TQS motif. TWIST1 TQS phosphorylation state determines the intensity of TWIST1-induced pro-metastatic ability in prostate cancer cells, which may be partly explained mechanistically by TWIST1 dimeric partner choice.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Protein Interaction Domains and Motifs , Twist-Related Protein 1/metabolism , Amino Acid Motifs , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cluster Analysis , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Heterografts , Humans , Male , Mutation , Neoplasm Metastasis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism , Transcriptome , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/genetics
4.
Mol Cancer Res ; 11(11): 1387-400, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23982216

ABSTRACT

UNLABELLED: Twist1, a basic helix-loop-helix transcription factor, plays a key role during development and is a master regulator of the epithelial-mesenchymal transition (EMT) that promotes cancer metastasis. Structure-function relationships of Twist1 to cancer-related phenotypes are underappreciated, so we studied the requirement of the conserved Twist box domain for metastatic phenotypes in prostate cancer. Evidence suggests that Twist1 is overexpressed in clinical specimens and correlated with aggressive/metastatic disease. Therefore, we examined a transactivation mutant, Twist1-F191G, in prostate cancer cells using in vitro assays, which mimic various stages of metastasis. Twist1 overexpression led to elevated cytoskeletal stiffness and cell traction forces at the migratory edge of cells based on biophysical single-cell measurements. Twist1 conferred additional cellular properties associated with cancer cell metastasis including increased migration, invasion, anoikis resistance, and anchorage-independent growth. The Twist box mutant was defective for these Twist1 phenotypes in vitro. Importantly, we observed a high frequency of Twist1-induced metastatic lung tumors and extrathoracic metastases in vivo using the experimental lung metastasis assay. The Twist box was required for prostate cancer cells to colonize metastatic lung lesions and extrathoracic metastases. Comparative genomic profiling revealed transcriptional programs directed by the Twist box that were associated with cancer progression, such as Hoxa9. Mechanistically, Twist1 bound to the Hoxa9 promoter and positively regulated Hoxa9 expression in prostate cancer cells. Finally, Hoxa9 was important for Twist1-induced cellular phenotypes associated with metastasis. These data suggest that the Twist box domain is required for Twist1 transcriptional programs and prostate cancer metastasis. IMPLICATIONS: Targeting the Twist box domain of Twist1 may effectively limit prostate cancer metastatic potential.


Subject(s)
Neoplasm Metastasis/genetics , Nuclear Proteins/metabolism , Prostatic Neoplasms/genetics , Protein Structure, Tertiary/genetics , Twist-Related Protein 1/metabolism , Amino Acid Substitution , Animals , Biomarkers, Tumor , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , Transcriptional Activation , Twist-Related Protein 1/genetics
5.
PLoS One ; 8(6): e65726, 2013.
Article in English | MEDLINE | ID: mdl-23762417

ABSTRACT

Sorafenib (SOR) is the only systemic agent known to improve survival for hepatocellular carcinoma (HCC). However, SOR prolongs survival by less than 3 months and does not alter symptomatic progression. To improve outcomes, several phase I-II trials are currently examining SOR with radiation (RT) for HCC utilizing heterogeneous concurrent and sequential treatment regimens. Our study provides preclinical data characterizing the effects of concurrent versus sequential RT-SOR on HCC cells both in vitro and in vivo. Concurrent and sequential RT-SOR regimens were tested for efficacy among 4 HCC cell lines in vitro by assessment of clonogenic survival, apoptosis, cell cycle distribution, and γ-H2AX foci formation. Results were confirmed in vivo by evaluating tumor growth delay and performing immunofluorescence staining in a hind-flank xenograft model. In vitro, concurrent RT-SOR produced radioprotection in 3 of 4 cell lines, whereas sequential RT-SOR produced decreased colony formation among all 4. Sequential RT-SOR increased apoptosis compared to RT alone, while concurrent RT-SOR did not. Sorafenib induced reassortment into less radiosensitive phases of the cell cycle through G1-S delay and cell cycle slowing. More double-strand breaks (DSBs) persisted 24 h post-irradiation for RT alone versus concurrent RT-SOR. In vivo, sequential RT-SOR produced the greatest tumor growth delay, while concurrent RT-SOR was similar to RT alone. More persistent DSBs were observed in xenografts treated with sequential RT-SOR or RT alone versus concurrent RT-SOR. Sequential RT-SOR additionally produced a greater reduction in xenograft tumor vascularity and mitotic index than either concurrent RT-SOR or RT alone. In conclusion, sequential RT-SOR demonstrates greater efficacy against HCC than concurrent RT-SOR both in vitro and in vivo. These results may have implications for clinical decision-making and prospective trial design.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/therapy , Combined Modality Therapy/methods , Gamma Rays/therapeutic use , Histones/genetics , Liver Neoplasms/therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Clinical Trials as Topic , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , Hindlimb/blood supply , Hindlimb/metabolism , Hindlimb/pathology , Histones/metabolism , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Nude , Neovascularization, Pathologic/prevention & control , Niacinamide/therapeutic use , Radiation Tolerance , Sorafenib , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Cancer Biol Ther ; 14(4): 347-56, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358469

ABSTRACT

Outcomes for poor-risk localized prostate cancers treated with radiation are still insufficient. Targeting the "non-oncogene" addiction or stress response machinery is an appealing strategy for cancer therapeutics. Heat-shock-protein-90 (Hsp90), an integral member of this machinery, is a molecular chaperone required for energy-driven stabilization and selective degradation of misfolded "client" proteins, that is commonly overexpressed in tumor cells. Hsp90 client proteins include critical components of pathways implicated in prostate cancer cell survival and radioresistance, such as androgen receptor signaling and the PI3K-Akt-mTOR pathway. We examined the effects of a novel non-geldanamycin Hsp90 inhibitor, AUY922, combined with radiation (RT) on two prostate cancer cell lines, Myc-CaP and PC3, using in vitro assays for clonogenic survival, apoptosis, cell cycle distribution, γ-H2AX foci kinetics and client protein expression in pathways important for prostate cancer survival and radioresistance. We then evaluated tumor growth delay and effects of the combined treatment (RT-AUY922) on the PI3K-Akt-mTOR and AR pathways in a hind-flank tumor graft model. We observed that AUY922 caused supra-additive radiosensitization in both cell lines at low nanomolar doses with enhancement ratios between 1.4-1.7 (p < 0.01). RT-AUY922 increased apoptotic cell death compared with either therapy alone, induced G 2-M arrest and produced marked changes in client protein expression. These results were confirmed in vivo, where RT-AUY922 combination therapy produced supra-additive tumor growth delay compared with either therapy by itself in Myc-CaP and PC3 tumor grafts (both p < 0.0001). Our data suggest that combined RT-AUY922 therapy exhibits promising activity against prostate cancer cells, which should be investigated in clinical studies.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Resorcinols/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Down-Regulation , G2 Phase/drug effects , Humans , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Random Allocation , Signal Transduction/drug effects , Signal Transduction/radiation effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...