Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Int J Cancer ; 152(7): 1399-1413, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36346110

ABSTRACT

The mitochondrion is a gatekeeper of apoptotic processes, and mediates drug resistance to several chemotherapy agents used to treat cancer. Neuroblastoma is a common solid cancer in young children with poor clinical outcomes following conventional chemotherapy. We sought druggable mitochondrial protein targets in neuroblastoma cells. Among mitochondria-associated gene targets, we found that high expression of the mitochondrial adenine nucleotide translocase 2 (SLC25A5/ANT2), was a strong predictor of poor neuroblastoma patient prognosis and contributed to a more malignant phenotype in pre-clinical models. Inhibiting this transporter with PENAO reduced cell viability in a panel of neuroblastoma cell lines in a TP53-status-dependant manner. We identified the histone deacetylase inhibitor, suberanilohydroxamic acid (SAHA), as the most effective drug in clinical use against mutant TP53 neuroblastoma cells. SAHA and PENAO synergistically reduced cell viability, and induced apoptosis, in neuroblastoma cells independent of TP53-status. The SAHA and PENAO drug combination significantly delayed tumour progression in pre-clinical neuroblastoma mouse models, suggesting that these clinically advanced inhibitors may be effective in treating the disease.


Subject(s)
Adenine Nucleotide Translocator 2 , Antineoplastic Agents , Histone Deacetylase Inhibitors , Hydroxamic Acids , Neuroblastoma , Animals , Mice , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Hydroxamic Acids/therapeutic use , Mitochondria/metabolism , Neuroblastoma/drug therapy , Vorinostat/pharmacology , Adenine Nucleotide Translocator 2/antagonists & inhibitors
2.
J Cancer Res Clin Oncol ; 145(9): 2241-2250, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31342168

ABSTRACT

PURPOSE: The tripartite motif (TRIM)16 acts as a tumour suppressor in both squamous cell carcinoma (SCC) and melanoma. TRIM16 is known to be secreted by keratinocytes, but no studies have been reported yet to assess the relationship between TRIM16 keratinocyte expression and melanoma development. METHODS: To study the role of TRIM16 in skin cancer development, we developed a keratinocyte TRIM16-specific knockout mouse model, and used the classical two-stage skin carcinogenesis challenge method, to assess the loss of keratinocyte TRIM16 on both papilloma, SCC and melanoma development in the skin after topical carcinogen treatment. RESULTS: Heterozygous, but not homozygous, TRIM16 knockout mice exhibited an accelerated development of skin papillomas and melanomas, larger melanoma lesions and an increased potential for lymph node metastasis. CONCLUSION: This study provides the first evidence that keratinocyte loss of the putative melanoma tumour suppressor protein, TRIM16, enhances melanomagenesis. Our data also suggest that TRIM16 expression in keratinocytes is involved in cross talk between keratinocytes and melanocytes, and has a role in melanoma tumorigenesis.


Subject(s)
Carrier Proteins/genetics , Keratinocytes/metabolism , Loss of Heterozygosity/physiology , Lymph Nodes/metabolism , Melanocytes/metabolism , Melanoma/genetics , Skin Neoplasms/genetics , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carrier Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Gene Expression Regulation, Neoplastic , Keratinocytes/pathology , Lymph Nodes/pathology , Lymphatic Metastasis , Melanocytes/pathology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Knockout , Skin/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
3.
Oncotarget ; 7(32): 52166-52178, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27447557

ABSTRACT

There is an urgent need for better therapeutic options for advanced melanoma patients, particularly those without the BRAFV600E/K mutation. In melanoma cells, loss of TRIM16 expression is a marker of cell migration and metastasis, while the BRAF inhibitor, vemurafenib, induces melanoma cell growth arrest in a TRIM16-dependent manner. Here we identify a novel small molecule compound which sensitized BRAF wild-type melanoma cells to vemurafenib. High throughput, cell-based, chemical library screening identified a compound (C012) which significantly reduced melanoma cell viability, with limited toxicity for normal human fibroblasts. When combined with the BRAFV600E/K inhibitor, vemurafenib, C012 synergistically increased vemurafenib potency in 5 BRAFWT and 4 out of 5 BRAFV600E human melanoma cell lines (Combination Index: CI < 1), and, dramatically reduced colony forming ability. In addition, this drug combination was significantly anti-tumorigenic in vivo in a melanoma xenograft mouse model. The combination of vemurafenib and C012 markedly increased expression of TRIM16 protein, and knockdown of TRIM16 significantly reduced the growth inhibitory effects of the vemurafenib and C012 combination. These findings suggest that the combination of C012 and vemurafenib may have therapeutic potential for the treatment of melanoma, and, that reactivation of TRIM16 may be an effective strategy for patients with this disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , DNA-Binding Proteins/biosynthesis , Melanoma/pathology , Transcription Factors/biosynthesis , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Indoles/pharmacology , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Sulfonamides/pharmacology , Transcription Factors/drug effects , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Vemurafenib , Xenograft Model Antitumor Assays
4.
Molecules ; 21(7)2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27428941

ABSTRACT

Identification of the novel (E)-N'-((2-chloro-7-methoxyquinolin-3-yl)methylene)-3-(phenylthio)propanehydrazide scaffold 18 has led to the development of a new series of biologically active hydrazide compounds. The parent compound 18 and new quinoline derivatives 19-26 were prepared from the corresponding quinoline hydrazones and substituted carboxylic acids using EDC-mediated peptide coupling reactions. Further modification of the parent compound 18 was achieved by replacement of the quinoline moiety with other aromatic systems. All the newly synthesized compounds were evaluated for their anti-cancer activity against the SH-SY5Y and Kelly neuroblastoma cell lines, as well as the MDA-MB-231 and MCF-7 breast adenocarcinoma cell lines. Analogues 19 and 22 significantly reduced the cell viability of neuroblastoma cancer cells with micromolar potency and significant selectivity over normal cells. The quinoline hydrazide 22 also induced G1 cell cycle arrest, as well as upregulation of the p27(kip1) cell cycle regulating protein.


Subject(s)
Hydrazines/chemical synthesis , Hydrazines/pharmacology , Quinolines/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Fibroblasts/drug effects , Humans , Hydrazines/chemistry , Hydrazones/chemistry , Molecular Structure , Structure-Activity Relationship
5.
Mol Oncol ; 10(6): 866-78, 2016 06.
Article in English | MEDLINE | ID: mdl-26996379

ABSTRACT

The MYCN gene is amplified and overexpressed in a large proportion of high stage neuroblastoma patients and has been identified as a key driver of tumorigenesis. However, the mechanism by which MYCN promotes tumor initiation is poorly understood. Here we conducted metabolic profiling of pre-malignant sympathetic ganglia and tumors derived from the TH-MYCN mouse model of neuroblastoma, compared to non-malignant ganglia from wildtype littermates. We found that metabolites involved in the biosynthesis of glutathione, the most abundant cellular antioxidant, were the most significantly upregulated metabolic pathway at tumor initiation, and progressively increased to meet the demands of tumorigenesis. A corresponding increase in the expression of genes involved in ribosomal biogenesis suggested that MYCN-driven transactivation of the protein biosynthetic machinery generated the necessary substrates to drive glutathione biosynthesis. Pre-malignant sympathetic ganglia from TH-MYCN mice had higher antioxidant capacity and required glutathione upregulation for cell survival, when compared to wildtype ganglia. Moreover, in vivo administration of inhibitors of glutathione biosynthesis significantly delayed tumorigenesis when administered prophylactically and potentiated the anticancer activity of cytotoxic chemotherapy against established tumors. Together these results identify enhanced glutathione biosynthesis as a selective metabolic adaptation required for initiation of MYCN-driven neuroblastoma, and suggest that glutathione-targeted agents may be used as a potential preventative strategy, or as an adjuvant to existing chemotherapies in established disease.


Subject(s)
Carcinogenesis/metabolism , Ganglia, Sympathetic/pathology , Glutathione/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Peripheral Nervous System Neoplasms/metabolism , Animals , Biosynthetic Pathways , Carcinogenesis/pathology , Disease Models, Animal , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/metabolism , Humans , Metabolome , Mice , Mice, Transgenic , Neuroblastoma/pathology , Peripheral Nervous System Neoplasms/pathology
6.
Sci Transl Med ; 7(312): 312ra176, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26537256

ABSTRACT

Amplification of the MYCN oncogene predicts treatment resistance in childhood neuroblastoma. We used a MYC target gene signature that predicts poor neuroblastoma prognosis to identify the histone chaperone FACT (facilitates chromatin transcription) as a crucial mediator of the MYC signal and a therapeutic target in the disease. FACT and MYCN expression created a forward feedback loop in neuroblastoma cells that was essential for maintaining mutual high expression. FACT inhibition by the small-molecule curaxin compound CBL0137 markedly reduced tumor initiation and progression in vivo. CBL0137 exhibited strong synergy with standard chemotherapy by blocking repair of DNA damage caused by genotoxic drugs, thus creating a synthetic lethal environment in MYCN-amplified neuroblastoma cells and suggesting a treatment strategy for MYCN-driven neuroblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , High Mobility Group Proteins/antagonists & inhibitors , Nervous System Neoplasms/drug therapy , Nervous System Neoplasms/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcriptional Elongation Factors/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Carbazoles/therapeutic use , DNA Repair/drug effects , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Humans , Molecular Targeted Therapy , Signal Transduction/drug effects , Transcriptional Elongation Factors/metabolism
7.
Mol Oncol ; 9(7): 1484-500, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25963741

ABSTRACT

Retinoids are an important component of neuroblastoma therapy at the stage of minimal residual disease, yet 40-50% of patients treated with 13-cis-retinoic acid (13-cis-RA) still relapse, indicating the need for more effective retinoid therapy. Vorinostat, or Suberoylanilide hydroxamic acid (SAHA), is a potent inhibitor of histone deacetylase (HDAC) classes I & II and has antitumor activity in vitro and in vivo. Fenretinide (4-HPR) is a synthetic retinoid which acts on cancer cells through both nuclear retinoid receptor and non-receptor mechanisms. In this study, we found that the combination of 4-HPR + SAHA exhibited potent cytotoxic effects on neuroblastoma cells, much more effective than 13-cis-RA + SAHA. The 4-HPR + SAHA combination induced caspase-dependent apoptosis through activation of caspase 3, reduced colony formation and cell migration in vitro, and tumorigenicity in vivo. The 4-HPR and SAHA combination significantly increased mRNA expression of thymosin-beta-4 (Tß4) and decreased mRNA expression of retinoic acid receptor α (RARα). Importantly, the up-regulation of Tß4 and down-regulation of RARα were both necessary for the 4-HPR + SAHA cytotoxic effect on neuroblastoma cells. Moreover, Tß4 knockdown in neuroblastoma cells increased cell migration and blocked the effect of 4-HPR + SAHA on cell migration and focal adhesion formation. In primary human neuroblastoma tumor tissues, low expression of Tß4 was associated with metastatic disease and predicted poor patient prognosis. Our findings demonstrate that Tß4 is a novel therapeutic target in neuroblastoma, and that 4-HPR + SAHA is a potential therapy for the disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neuroblastoma/drug therapy , Thymosin/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Fenretinide/administration & dosage , Humans , Hydroxamic Acids/administration & dosage , Neuroblastoma/metabolism , Neuroblastoma/pathology , Thymosin/genetics , Vorinostat
8.
Oncotarget ; 5(20): 10127-39, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25333256

ABSTRACT

High basal or induced expression of the tripartite motif protein, TRIM16, leads to reduce cell growth and migration of neuroblastoma and skin squamous cell carcinoma cells. However, the role of TRIM16 in melanoma is currently unknown. TRIM16 protein levels were markedly reduced in human melanoma cell lines, compared with normal human epidermal melanocytes due to both DNA methylation and reduced protein stability. TRIM16 knockdown strongly increased cell migration in normal human epidermal melanocytes, while TRIM16 overexpression reduced cell migration and proliferation of melanoma cells in an interferon beta 1 (IFNß1)-dependent manner. Chromatin immunoprecipitation assays revealed TRIM16 directly bound the IFNß1 gene promoter. Low level TRIM16 expression in 91 melanoma patient samples, strongly correlated with lymph node metastasis, and, predicted poor patient prognosis in a separate cohort of 170 melanoma patients with lymph node metastasis. The BRAF inhibitor, vemurafenib, increased TRIM16 protein levels in melanoma cells in vitro, and induced growth arrest in BRAF-mutant melanoma cells in a TRIM16-dependent manner. High levels of TRIM16 in melanoma tissues from patients treated with Vemurafenib correlated with clinical response. Our data, for the first time, demonstrates TRIM16 is a marker of cell migration and metastasis, and a novel treatment target in melanoma.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon-beta/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Humans , Indoles/pharmacology , Interferon-beta/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Promoter Regions, Genetic , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Sulfonamides/pharmacology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Vemurafenib
9.
J Pathol ; 226(3): 451-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22009481

ABSTRACT

Retinoid therapy is used for chemo-prevention in immuno-suppressed patients at high risk of developing skin cancer. The retinoid signalling molecule, tripartite motif protein 16 (TRIM16), is a regulator of keratinocyte differentiation and a tumour suppressor in retinoid-sensitive neuroblastoma. We sought to determine the role of TRIM16 in skin squamous cell carcinoma (SCC) pathogenesis. We have shown that TRIM16 expression was markedly reduced during the histological progression from normal skin to actinic keratosis and SCC. SCC cell lines exhibited lower cytoplasmic and nuclear TRIM16 expression compared with primary human keratinocyte (PHK) cells due to reduced TRIM16 protein stability. Overexpressed TRIM16 translocated to the nucleus, inducing growth arrest and cell differentiation. In SCC cells, TRIM16 bound to and down regulated nuclear E2F1, this is required for cell replication. Retinoid treatment increased nuclear TRIM16 expression in retinoid-sensitive PHK cells, but not in retinoid-resistant SCC cells. Overexpression of TRIM16 reduced SCC cell migration, which required the C-terminal RET finger protein (RFP)-like domain of TRIM16. The mesenchymal intermediate filament protein, vimentin, was directly bound and down-regulated by TRIM16 and was required for TRIM16-reduced cell migration. Taken together, our data suggest that loss of TRIM16 expression plays an important role in the development of cutaneous SCC and is a determinant of retinoid sensitivity.


Subject(s)
Carcinoma, Squamous Cell/etiology , DNA-Binding Proteins/metabolism , Skin Neoplasms/etiology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Movement/physiology , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Dermatologic Agents/pharmacology , Down-Regulation , Humans , Immunohistochemistry , In Vitro Techniques , Isotretinoin/pharmacology , Protein Binding , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Tripartite Motif Proteins , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , Vimentin/metabolism
10.
J Clin Endocrinol Metab ; 94(12): 4703-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19875482

ABSTRACT

CONTEXT: GH abuse is a significant problem in many sports, and there is currently no robust test that allows detection of doping beyond a short window after administration. OBJECTIVE: Our objective was to evaluate gene expression profiling in peripheral blood leukocytes in-vivo as a test for GH doping in humans. DESIGN: Seven men and thirteen women were administered GH, 2 mg/d sc for 8 wk. Blood was collected at baseline and at 8 wk. RNA was extracted from the white cell fraction. Microarray analysis was undertaken using Agilent 44K G4112F arrays using a two-color design. Quantitative RT-PCR using TaqMan gene expression assays was performed for validation of selected differentially expressed genes. RESULTS: GH induced an approximately 2-fold increase in circulating IGF-I that was maintained throughout the 8 wk of the study. GH induced significant changes in gene expression with 353 in women and 41 in men detected with a false discovery rate of less than 5%. None of the differentially expressed genes were common between men and women. The maximal changes were a doubling for up-regulated or halving for down-regulated genes, similar in magnitude to the variation between individuals. Quantitative RT-PCR for seven target genes showed good concordance between microarray and quantitative PCR data in women but not in men. CONCLUSION: Gene expression analysis of peripheral blood leukocytes is unlikely to be a viable approach for the detection of GH doping.


Subject(s)
Doping in Sports/methods , Gene Expression/drug effects , Human Growth Hormone/pharmacology , Recombinant Proteins/pharmacology , Adult , Double-Blind Method , Female , Gene Expression Profiling , Humans , Insulin-Like Growth Factor I/metabolism , Male , Oligonucleotide Array Sequence Analysis , RNA/biosynthesis , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Sex Characteristics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL