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.
Oncogene ; 40(24): 4214-4228, 2021 06.
Article in English | MEDLINE | ID: mdl-34079088

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is often being diagnosed at an advanced stage, conferring a poor prognosis. The probability of local tumor control after radiotherapy depends on the eradication of cancer stem cells (CSCs) with activated DNA repair. This study provides evidence that the CSC-related transcription factor Oct4 contributes to HNSCC radioresistance by regulating DNA damage response and the CSC phenotype. Knockdown of Oct4 A isoform reduced self-renewal capacity in HNSCC and led to partial tumor cell radiosensitization caused by transcriptional downregulation of the cell cycle checkpoint kinases CHK1 and WEE1 and homologous recombination (HR) repair genes PSMC3IP and RAD54L. Besides, PARP inhibition with Olaparib selectively radiosensitized Oct4 A knockout, but not wild-type HNSCC cells. This finding links Oct4 A to the HR-mediated DNA repair mechanisms. In turn, knockdown of PSMC3IP and RAD54L reduced the HNSCC self-renewal capacity and clonogenic cell survival after irradiation, suggesting the interplay between DNA repair and the CSC phenotype. Similar to the effect of Oct4 knockdown, overexpression of Oct4 also resulted in significant HNSCC radiosensitization and increased DNA damage, suggesting that Oct4-dependent regulation of DNA repair depends on its fine-tuned expression. In line with this observation, HNSCC patients with high and low nuclear Oct4 expression at the invasive tumor front exhibited better loco-regional tumor control after postoperative radio(chemo)therapy compared to the intermediate expression subgroup. Thus, we found that the Oct4-driven transcriptional program plays a critical role in regulating HNSCC radioresistance, and a combination of radiotherapy with PARP inhibitors may induce synthetic lethality in Oct4-deregulated tumors.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Head and Neck Neoplasms/genetics , Homologous Recombination/genetics , Neoplastic Stem Cells/pathology , Nuclear Proteins/genetics , Octamer Transcription Factor-3/genetics , Radiation Tolerance/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Trans-Activators/genetics , Adult , Aged , DNA Damage/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Squamous Cell Carcinoma of Head and Neck/pathology , Young Adult
2.
Clin Cancer Res ; 25(10): 3152-3163, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30670494

ABSTRACT

PURPOSE: The heavy chain of the CD98 protein (CD98hc) is encoded by the SLC3A2 gene. Together with the light subunit LAT1, CD98hc constitutes a heterodimeric transmembrane amino acid transporter. High SLC3A2 mRNA expression levels are associated with poor prognosis in patients with head and neck squamous cell carcinoma (HNSCC) treated with radiochemotherapy. Little is known regarding the CD98hc protein-mediated molecular mechanisms of tumor radioresistance. EXPERIMENTAL DESIGN: CD98hc protein expression levels were correlated with corresponding tumor control dose 50 (TCD50) in HNSCC xenograft models. Expression levels of CD98hc and LAT1 in HNSCC cells were modulated by siRNA or CRISPR/Cas9 gene editing. HNSCC cell phenotypes were characterized by transcription profiling, plasma membrane proteomics, metabolic analysis, and signaling pathway activation. Expression levels of CD98hc and LAT1 proteins were examined by IHC analysis of tumor tissues from patients with locally advanced HNSCC treated with primary radiochemotherapy (RCTx). Primary endpoint was locoregional tumor control (LRC). RESULTS: High expression levels of CD98hc resulted in an increase in mTOR pathway activation, amino acid metabolism, and DNA repair as well as downregulation of oxidative stress and autophagy. High expression levels of CD98hc and LAT1 proteins were significantly correlated and associated with an increase in radioresistance in HNSCC in vitro and in vivo models. High expression of both proteins identified a poor prognosis subgroup in patients with locally advanced HNSCC after RCTx. CONCLUSIONS: We found that CD98hc-associated signaling mechanisms play a central role in the regulation of HNSCC radioresistance and may be a promising target for tumor radiosensitization.


Subject(s)
Fusion Regulatory Protein 1, Heavy Chain/genetics , Radiation Tolerance/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Amino Acids/metabolism , Biological Transport , Biomarkers, Tumor , Cell Line, Tumor , Chemoradiotherapy , Citric Acid Cycle , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Gene Expression , Gene Knockdown Techniques , Humans , Immunohistochemistry , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Oxidative Stress/genetics , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/radiotherapy
3.
Cancer Res ; 76(9): 2637-51, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26984757

ABSTRACT

Radiotherapy is a mainstay of curative prostate cancer treatment, but risks of recurrence after treatment remain significant in locally advanced disease. Given that tumor relapse can be attributed to a population of cancer stem cells (CSC) that survives radiotherapy, analysis of this cell population might illuminate tactics to personalize treatment. However, this direction remains challenging given the plastic nature of prostate cancers following treatment. We show here that irradiating prostate cancer cells stimulates a durable upregulation of stem cell markers that epigenetically reprogram these cells. In both tumorigenic and radioresistant cell populations, a phenotypic switch occurred during a course of radiotherapy that was associated with stable genetic and epigenetic changes. Specifically, we found that irradiation triggered histone H3 methylation at the promoter of the CSC marker aldehyde dehydrogenase 1A1 (ALDH1A1), stimulating its gene transcription. Inhibiting this methylation event triggered apoptosis, promoted radiosensitization, and hindered tumorigenicity of radioresistant prostate cancer cells. Overall, our results suggest that epigenetic therapies may restore the cytotoxic effects of irradiation in radioresistant CSC populations. Cancer Res; 76(9); 2637-51. ©2016 AACR.


Subject(s)
Epigenesis, Genetic/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Prostatic Neoplasms/genetics , Radiation Tolerance/genetics , Retinal Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Comparative Genomic Hybridization , DNA Methylation/radiation effects , Flow Cytometry , Heterografts , Histones/genetics , Histones/radiation effects , Humans , Male , Mice , Mice, Nude , Microscopy, Fluorescence , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/radiation effects , Radiotherapy , Retinal Dehydrogenase/radiation effects
4.
Anticancer Agents Med Chem ; 16(1): 38-58, 2016.
Article in English | MEDLINE | ID: mdl-26179271

ABSTRACT

Recent discoveries have provided the compelling evidence that stem cell populations within each individual tumor are key contributors of therapy failure regardless of whether these populations are transient or stable. Thus, it is becoming increasingly clear that efficient tumor treatment requires eradication of the entire CSC population. The potential role of CSCs in tumor initiation and relapse has motivated an investigation of the CSCspecific treatments. However, development of the therapeutic strategies targeting CSCs might be challenged by a high diversity and plasticity of CSC features. Moreover, taking in account that an origin of CSC remains controversial and accumulating experimental evidence suggests a possibility of tumor cell reprogramming, efficient anti-cancer treatment should eradicate both CSCs and tumor bulk as well as prevent tumor dedifferentiation. In this article we discuss new insights into the stem cell concept of tumor development, review the treatment strategies eradicating CSCs that have been evaluated in preclinical and clinical studies and summarize the strategies to identify new CSC-targeted therapy.


Subject(s)
Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Humans , Neoplastic Stem Cells/pathology
5.
Oncotarget ; 6(33): 34494-509, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26460734

ABSTRACT

Despite recent advances in understanding of the molecular pathogenesis and improvement of treatment techniques, locally advanced head and neck squamous cell carcinoma (HNSCC) remains associated with an unfavorable prognosis. Compelling evidence suggests that cancer stem cells (CSC) may cause tumor recurrence if they are not eradicated by current therapies as radiotherapy or radio-chemotherapy. Recent in vitro studies have demonstrated that CSCs may be protected from treatment-induced death by multiple intrinsic and extrinsic mechanisms. Therefore, early determination of CSC abundance in tumor biopsies prior-treatment and development of therapeutics, which specifically target CSCs, are promising strategies to optimize treatment. Here we provide evidence that aldehyde dehydrogenase (ALDH) activity is indicative for radioresistant HNSCC CSCs. Our study suggests that ALDH+ cells comprise a population that maintains its tumorigenic properties in vivo after irradiation and may provide tumor regrowth after therapy. We found that ALDH activity in HNSCC cells can be attributed, at least in part, to the ALDH1A3 isoform and inhibition of the ALDH1A3 expression by small interfering RNA (siRNA) decreases tumor cell radioresistance. The expression dynamic of ALDH1A3 upon irradiation by either induction or selection of the ALDH1A3 positive population correlates to in vivo curability, suggesting that changes in protein expression during radiotherapy are indicative for tumor radioresistance. Our data indicate that ALDH1A3+ HNSCC cells may contribute to tumor relapse after irradiation, and inhibition of this cell population might improve therapeutic response to radiotherapy.


Subject(s)
Aldehyde Oxidoreductases/metabolism , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Neoplastic Stem Cells/pathology , Radiation Tolerance/physiology , Animals , Biomarkers, Tumor/analysis , Blotting, Western , Carcinoma, Squamous Cell/enzymology , Cell Line, Tumor , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockdown Techniques , Head and Neck Neoplasms/enzymology , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Squamous Cell Carcinoma of Head and Neck , Tissue Array Analysis , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...