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.
EJHaem ; 1(2): 517-526, 2020 Nov.
Article in English | MEDLINE | ID: mdl-35845006

ABSTRACT

Follicular lymphoma (FL) is the most common indolent non-Hodgkin lymphoma. Twenty to twenty-five percent of FL patients have progression of disease within 24 months. These patients may benefit from immunotherapy if intact antigen presentation is present. Molecular mechanisms impairing major histocompatibility complex class-I (MHC-I) in FL remain undefined. Here, by sequencing of 172 FL tumours, we found the MHC-I transactivator NLRC5 was the most frequent gene abnormality in the MHC-I pathway. Pyrosequencing showed that epigenetic silencing of the NLRC5 promoter occurred in 30% of cases and was mutually exclusive to copy number loss (CNL) in NLRC5 (∼6% of cases). Hypermethylation and CNLs ("NLRC5 aberrant") had reduced NLRC5 gene expression compared to wild-type (WT) cases. By NanoString, there was reduced gene expression of the MHC-I pathway in aberrant tissues, including immunoproteasome components (PSMB8 and PSMB9), peptide transporters of antigen processing (TAP1), and MHC-I (HLA-A), compared to WT. By immunofluorescent microscopy, fewer NLRC5 protein-expressing malignant B-cells were observed in NLRC5 aberrant tissue sections compared to NLRC5 WT (P < .01). Consistent with a pivotal role in the activation of CD8+ T-cells, both CD8 and CD137 strongly correlated with NLRC5 expression (both r > 0.7; P < .0001). Further studies are required to determine whether patients with aberrant NLRC5 have a diminished response to immunotherapy.

2.
Sci Transl Med ; 10(447)2018 06 27.
Article in English | MEDLINE | ID: mdl-29950445

ABSTRACT

Patient mortality rates have remained stubbornly high (40%) for the past 35 years in head and neck squamous cell carcinoma (HNSCC) due to inherent or acquired drug resistance. Thus, a critical issue in advanced SCC is to identify and target the mechanisms that contribute to therapy resistance. We report that the transcriptional inhibitor, E2F7, is mislocalized to the cytoplasm in >80% of human HNSCCs, whereas the transcriptional activator, E2F1, retains localization to the nucleus in SCC. This results in an imbalance in the control of E2F-dependent targets such as SPHK1, which is derepressed and drives resistance to anthracyclines in HNSCC. Specifically, we show that (i) E2F7 is subject to exportin 1 (XPO1)-dependent nuclear export, (ii) E2F7 is selectively mislocalized in most of SCC and multiple other tumor types, (iii) mislocalization of E2F7 in HNSCC causes derepression of Sphk1 and drives anthracycline resistance, and (iv) anthracycline resistance can be reversed with a clinically available inhibitor of XPO1, selinexor, in xenotransplant models of HNSCC. Thus, we have identified a strategy to repurpose anthracyclines for use in SCC. More generally, we provide a strategy to restore the balance of E2F1 (activator) and E2F7 (inhibitor) activity in cancer.


Subject(s)
Anthracyclines/pharmacology , Cell Nucleus/metabolism , Drug Resistance, Neoplasm/drug effects , E2F7 Transcription Factor/metabolism , Karyopherins/antagonists & inhibitors , Molecular Targeted Therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , Doxorubicin/pharmacology , E2F1 Transcription Factor/metabolism , Humans , Karyopherins/metabolism , Mice, Inbred NOD , Mice, SCID , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
3.
J Invest Dermatol ; 135(10): 2446-2454, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25938557

ABSTRACT

Recent studies have reported that epigenetic mechanisms may regulate the initiation and progress of squamous differentiation in normal and transformed keratinocytes. In particular, the role of the repressive H3K27me3 mark in the regulation of squamous differentiation has been prominent. However, there is conflicting literature showing that squamous differentiation may be dependent upon or independent of changes in H3K27me3 status. In this study we have examined the binding of trimethylated H3K27 to the promoters of proliferation or differentiation genes in keratinocytes undergoing squamous differentiation in vitro and in vivo. Initially, we examined the expression levels for EZH1, EZH2, and H3K27me3 in differentiating keratinocytes in vitro and in vivo. We extended this to include H3K27me3 chromatin immunoprecipitation sequencing (ChIP-seq). Based on these studies, we could find no evidence for an association between widespread gain or loss of H3K27me3 on the promoters of proliferation-specific or differentiation-specific target genes, respectively, during squamous differentiation in adult human keratinocytes. These data suggest that squamous differentiation may occur independent of regulation by H3K27me3 on proliferation and differentiation genes of normal adult human keratinocytes.


Subject(s)
Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic , Adult , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Chromatin Immunoprecipitation , DNA Methylation , Enhancer of Zeste Homolog 2 Protein , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Sampling Studies , Skin Neoplasms/genetics , Skin Neoplasms/pathology
4.
Mol Cancer Ther ; 14(8): 1939-50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26018753

ABSTRACT

We have previously shown that E2F7 contributes to drug resistance in head and neck squamous cell carcinoma (HNSCC) cells. Considering that dysregulation of responses to chemotherapy-induced cytotoxicity is one of the major reasons for treatment failure in HNSCC, identifying the downstream effectors that regulate E2F7-dependent sensitivity to chemotherapeutic agents may have direct clinical impact. We used transcriptomic profiling to identify candidate pathways that contribute to E2F7-dependent resistance to doxorubicin. We then manipulated the expression of the candidate pathway using overexpression and knockdown in in vitro and in vivo models of SCC to demonstrate causality. In addition, we examined the expression of E2F7 and RacGAP1 in a custom tissue microarray (TMA) generated from HNSCC patient samples. Transcriptomic profiling identified RacGAP1 as a potential mediator of E2F7-dependent drug resistance. We validated E2F7-dependent upregulation of RacGAP1 in doxorubicin-insensitive SCC25 cells. Extending this, we found that selective upregulation of RacGAP1 induced doxorubicin resistance in previously sensitive KJDSV40. Similarly, stable knockdown of RacGAP1 in insensitive SCC25 cells induced sensitivity to doxorubicin in vitro and in vivo. RacGAP1 expression was validated in a TMA, and we showed that HNSCCs that overexpress RacGAP1 are associated with a poorer patient overall survival. Furthermore, E2F7-induced doxorubicin resistance was mediated via RacGAP1-dependent activation of AKT. Finally, we show that SCC cells deficient in RacGAP1 grow slower and are sensitized to the cytotoxic actions of doxorubicin in vivo. These findings identify RacGAP1 overexpression as a novel prognostic marker of survival and a potential target to sensitize SCC to doxorubicin.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , E2F7 Transcription Factor/metabolism , GTPase-Activating Proteins/genetics , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/mortality , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , E2F7 Transcription Factor/genetics , Female , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Guanosine Triphosphate/metabolism , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
5.
Clin Cancer Res ; 21(2): 417-27, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25411162

ABSTRACT

PURPOSE: Head and neck squamous cell carcinomas (HNSCC) are frequently drug resistant and have a mortality rate of 45%. We have previously shown that E2F7 may contribute to drug resistance in SCC cells. However, the mechanism and pathways involved remain unknown. EXPERIMENTAL DESIGN: We used transcriptomic profiling to identify candidate pathways that may contribute to E2F7-dependent resistance to anthracyclines. We then manipulated the activity/expression of the candidate pathway using overexpression, knockdown, and pharmacological inhibitors in in vitro and in vivo models of SCC to demonstrate causality. In addition, we examined the expression of E2F7 and a downstream effector in a tissue microarray (TMA) generated from HNSCC patient samples. RESULTS: E2F7-deficient keratinocytes were selectively sensitive to doxorubicin and this was reversed by overexpressing E2F7. Transcriptomic profiling identified Sphingosine kinase 1 (Sphk1) as a potential mediator of E2F7-dependent drug resistance. Knockdown and overexpression studies revealed that Sphk1 was a downstream target of E2F7. TMA studies showed that E2F7 overexpression correlated with Sphk1 overexpression in human HNSCC. Moreover, inhibition of Sphk1 by shRNA or the Sphk1-specific inhibitor, SK1-I (BML-EI411), enhanced the sensitivity of SCC cells to doxorubicin in vitro and in vivo. Furthermore, E2F7-induced doxorubicin resistance was mediated via Sphk1-dependent activation of AKT in vitro and in vivo. CONCLUSION: We identify a novel drugable pathway in which E2F7 directly increases the transcription and activity of the Sphk1/S1P axis resulting in activation of AKT and subsequent drug resistance. Collectively, this novel combinatorial therapy can potentially be trialed in humans using existing agents.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Doxorubicin/pharmacology , E2F7 Transcription Factor/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , HEK293 Cells , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-akt/metabolism , Xenograft Model Antitumor Assays
6.
Mol Cancer ; 11: 74, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-23021083

ABSTRACT

BACKGROUND: Overexpression of CEACAM6 has been reported for a number of malignancies. However, the mechanism of how CEACAM6 contributes to cancer formation and its role in head and neck squamous cell carcinoma (HNSCC) remains unclear. Therefore, we examined the role of CEACAM6 in head and neck squamous cell carcinoma (HNSCC). METHODS: CEACAM6 expression was examined in normal squamous epithelia as well as a number of patient HNSCC samples and tumours derived from HNSCC cell lines injected into NOD/SCID mice. CEACAM6 expression was manipulated in HNSCC cell lines by shRNA-mediated CEACAM6 knockdown or virally-delivered overexpression of CEACAM6. The role of CEACAM6 in tumour growth and chemotherapeutic sensitivity was then assessed in vivo and in vitro respectively. RESULTS: CEACAM6 expression was significantly increased in highly tumourigenic HNSCC cell lines when compared to poorly tumourigenic HNSCC cell lines. Moreover, HNSCC patient tumours demonstrated focal expression of CEACAM6. Functional investigation of CEACAM6, involving over-expression and knock down studies, demonstrated that CEACAM6 over-expression could enhance tumour initiating activity and tumour growth via activation of AKT and suppression of caspase-3 mediated cell death. CONCLUSION: We report that CEACAM6 is focally overexpressed in a large fraction of human HNSCCs in situ. We also show that over-expression of CEACAM6 increases tumour growth and tumour initiating activity by suppressing PI3K/AKT-dependent apoptosis of HNSCC in a xenotransplant model of HNSCC. Finally, our studies indicate that foci of CEACAM6 expressing cells are selectively ablated by treatment of xenotransplant tumours with pharmacological inhibitors of PI3K/AKT in vivo.


Subject(s)
Antigens, CD/genetics , Apoptosis/genetics , Carcinoma, Squamous Cell/genetics , Cell Adhesion Molecules/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression , Head and Neck Neoplasms/genetics , Animals , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , GPI-Linked Proteins/genetics , Gene Knockdown Techniques , Head and Neck Neoplasms/pathology , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/administration & dosage , Quinolines/pharmacology , Squamous Cell Carcinoma of Head and Neck
SELECTION OF CITATIONS
SEARCH DETAIL
...