Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 23(9)2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35563182

ABSTRACT

The progression of anchorage-dependent epithelial cells to anchorage-independent growth represents a critical hallmark of malignant transformation. Using an in vitro model of human papillomavirus (HPV)-induced transformation, we previously showed that acquisition of anchorage-independent growth is associated with marked (epi)genetic changes, including altered expression of microRNAs. However, the laborious nature of the conventional growth method in soft agar to measure this phenotype hampers a high-throughput analysis. We developed alternative functional screening methods using 96- and 384-well ultra-low attachment plates to systematically investigate microRNAs regulating anchorage-independent growth. SiHa cervical cancer cells were transfected with a microRNA mimic library (n = 2019) and evaluated for cell viability. We identified 84 microRNAs that consistently suppressed growth in three independent experiments. Further validation in three cell lines and comparison of growth in adherent and ultra-low attachment plates yielded 40 microRNAs that specifically reduced anchorage-independent growth. In conclusion, ultra-low attachment plates are a promising alternative for soft-agar assays to study anchorage-independent growth and are suitable for high-throughput functional screening. Anchorage independence suppressing microRNAs identified through our screen were successfully validated in three cell lines. These microRNAs may provide specific biomarkers for detecting and treating HPV-induced precancerous lesions progressing to invasive cancer, the most critical stage during cervical cancer development.


Subject(s)
Alphapapillomavirus , MicroRNAs , Papillomavirus Infections , Uterine Cervical Neoplasms , Agar , Alphapapillomavirus/genetics , Cell Transformation, Neoplastic/genetics , Female , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Papillomaviridae/genetics , Papillomavirus Infections/metabolism , Uterine Cervical Neoplasms/pathology
2.
Mol Cancer Ther ; 16(3): 540-550, 2017 03.
Article in English | MEDLINE | ID: mdl-27980104

ABSTRACT

Patients with advanced stage head and neck squamous cell carcinoma (HNSCC) are often treated with cisplatin-containing chemoradiation protocols. Although cisplatin is an effective radiation sensitizer, it causes severe toxicity and not all patients benefit from the combination treatment. HNSCCs expectedly not responding to cisplatin may better be treated with surgery and postoperative radiation or cetuximab and radiation, but biomarkers to personalize chemoradiotherapy are not available. We performed an unbiased genome-wide functional genetic screen in vitro to identify genes that influence the response to cisplatin in HNSCC cells. By siRNA-mediated knockdown, we identified the Fanconi anemia/BRCA pathway as the predominant pathway for cisplatin response in HNSCC cells. We also identified the involvement of the SHFM1 gene in the process of DNA cross-link repair. Furthermore, expression profiles based on these genes predict the prognosis of radiation- and chemoradiation-treated head and neck cancer patients. This genome-wide functional analysis designated the genes that are important in the response of HNSCC to cisplatin and may guide further biomarker validation. Cisplatin imaging as well as biomarkers that indicate the activity of the Fanconi anemia/BRCA pathway in the tumors are the prime candidates. Mol Cancer Ther; 16(3); 540-50. ©2016 AACR.


Subject(s)
Cisplatin/pharmacology , Fanconi Anemia Complementation Group Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Genome-Wide Association Study , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Signal Transduction , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Cell Cycle/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Genomics/methods , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , RNA, Small Interfering/genetics , Squamous Cell Carcinoma of Head and Neck
3.
Nat Commun ; 6: 8399, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26423134

ABSTRACT

Warsaw breakage syndrome (WABS) is caused by defective DDX11, a DNA helicase that is essential for chromatid cohesion. Here, a paired genome-wide siRNA screen in patient-derived cell lines reveals that WABS cells do not tolerate partial depletion of individual APC/C subunits or the spindle checkpoint inhibitor p31(comet). A combination of reduced cohesion and impaired APC/C function also leads to fatal mitotic arrest in diploid RPE1 cells. Moreover, WABS cell lines, and several cancer cell lines with cohesion defects, display a highly increased response to a new cell-permeable APC/C inhibitor, apcin, but not to the spindle poison paclitaxel. Synthetic lethality of APC/C inhibition and cohesion defects strictly depends on a functional mitotic spindle checkpoint as well as on intact microtubule pulling forces. This indicates that the underlying mechanism involves cohesion fatigue in response to mitotic delay, leading to spindle checkpoint re-activation and lethal mitotic arrest. Our results point to APC/C inhibitors as promising therapeutic agents targeting cohesion-defective cancers.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/physiology , Sister Chromatid Exchange/physiology , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chromosome Segregation , Humans , Mitosis/physiology , Morpholines/pharmacology , Nocodazole/pharmacology , Paclitaxel/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sister Chromatid Exchange/drug effects , Tubulin Modulators/pharmacology
4.
Mol Cancer Ther ; 14(6): 1434-44, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25808837

ABSTRACT

Lung cancer is the most common cancer worldwide and on top of that has a very poor prognosis, which is reflected by a 5-year survival rate of 5% to 15%. Radiotherapy is an integral part of most treatment regimens for this type of tumor, often combined with radiosensitizing cytotoxic drugs. In this study, we identified many genes that could potentially be exploited for targeted radiosensitization using a genome-wide siRNA screen in non-small cell lung cancer (NSCLC) cells. The screen identified 433 siRNAs that potentially sensitize lung cancer cells to radiation. Validation experiments showed that knockdown of expression of Forkhead box M1 (FOXM1) or microtubule-associated serine/threonine kinase-like (MASTL) indeed causes radiosensitization in a panel of NSCLC cells. Strikingly, this effect was not observed in primary human fibroblasts, suggesting that the observed radiosensitization is specific for cancer cells. Phosphoproteomics analyses with and without irradiation showed that a number of cell-cycle-related proteins were significantly less phosphorylated after MASTL knockdown in comparison to the control, while there were no changes in the levels of phosphorylation of DNA damage response proteins. Subsequent analyses showed that MASTL knockdown cells respond differently to radiation, with a significantly shortened G2-M phase arrest and defects in cytokinesis, which are followed by a cell-cycle arrest. In summary, we have identified many potential therapeutic targets that could be used for radiosensitization of NSCLC cells, with MASTL being a very promising and druggable target to combine with radiotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Forkhead Transcription Factors/genetics , Genome, Human/genetics , Lung Neoplasms/radiotherapy , Microtubule-Associated Proteins/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Animals , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Line, Tumor , Down-Regulation/genetics , Down-Regulation/radiation effects , Forkhead Box Protein M1 , Forkhead Transcription Factors/metabolism , Gamma Rays , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , Phosphorylation/genetics , Phosphorylation/radiation effects , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 19(8): 1994-2003, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23444224

ABSTRACT

PURPOSE: Despite continuous improvement of treatment regimes, the mortality rates for non-small cell lung cancer (NSCLC) and head and neck squamous cell carcinoma (HNSCC) remain disappointingly high and novel anticancer agents are urgently awaited. EXPERIMENTAL DESIGN: We combined the data from genome-wide siRNA screens on tumor cell lethality in a lung and a head and neck cancer cell line. RESULTS: We identified 71 target genes that seem essential for the survival of both cancer types. We identified a cluster of 20 genes that play an important role during G2-M phase transition, underlining the importance of this cell-cycle checkpoint for tumor cell survival. Five genes from this cluster (CKAP5, KPNB1, RAN, TPX2, and KIF11) were evaluated in more detail and have been shown to be essential for tumor cell survival in both tumor types, but most particularly in HNSCC. Phenotypes that were observed following siRNA-mediated knockdown of KIF11 (kinesin family member 11) were reproduced by inhibition of KIF11 using the small-molecule inhibitor ispinesib (SB-715992). We showed that ispinesib induces a G2 arrest, causes aberrant chromosome segregation, and induces cell death in HNSCC in vitro, whereas primary keratinocytes are less sensitive. Furthermore, growth of HNSCC cells engrafted in immunodeficient mice was significantly inhibited after ispinesib treatment. CONCLUSION: This study identified a wide array of druggable genes for both lung and head and neck cancer. In particular, multiple genes involved in the G2-M checkpoint were shown to be essential for tumor cell survival, indicating their potential as anticancer targets.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Lung Neoplasms/genetics , RNA Interference , Animals , Benzamides/pharmacology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/prevention & control , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Genes, Essential/genetics , Genome-Wide Association Study/methods , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/prevention & control , Humans , Kinesins/genetics , Kinesins/metabolism , Lung Neoplasms/pathology , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Quinazolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , beta Karyopherins/genetics , beta Karyopherins/metabolism , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...