Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Br J Dermatol ; 186(4): 694-704, 2022 04.
Article in English | MEDLINE | ID: mdl-34773645

ABSTRACT

BACKGROUND: For patients with early American Joint Committee on Cancer (AJCC)-stage melanoma the combined loss of the autophagy regulatory protein AMBRA1 and the terminal differentiation marker loricrin in the peritumoral epidermis is associated with a significantly increased risk of metastasis. OBJECTIVES: The aim of the present study was to evaluate the potential contribution of melanoma paracrine transforming growth factor (TGF)-ß signalling to the loss of AMBRA1 in the epidermis overlying the primary tumour and disruption of epidermal integrity. METHODS: Immunohistochemistry was used to analyse AMBRA1 and TGF-ß2 in a cohort of 109 AJCC all-stage melanomas, and TGF-ß2 and claudin-1 in a cohort of 30 or 42 AJCC stage I melanomas, respectively, with known AMBRA1 and loricrin (AMLo) expression. Evidence of pre-ulceration was analysed in a cohort of 42 melanomas, with TGF-ß2 signalling evaluated in primary keratinocytes. RESULTS: Increased tumoral TGF-ß2 was significantly associated with loss of peritumoral AMBRA1 (P < 0·05), ulceration (P < 0·001), AMLo high-risk status (P < 0·05) and metastasis (P < 0·01). TGF-ß2 treatment of keratinocytes resulted in downregulation of AMBRA1, loricrin and claudin-1, while knockdown of AMBRA1 was associated with decreased expression of claudin-1 and increased proliferation of keratinocytes (P < 0·05). Importantly, we show loss of AMBRA1 in the peritumoral epidermis was associated with decreased claudin-1 expression (P < 0·05), parakeratosis (P < 0·01) and cleft formation in the dermoepidermal junction (P < 0·05). CONCLUSIONS: Collectively, these data suggest a paracrine mechanism whereby TGF-ß2 causes loss of AMBRA1 overlying high-risk AJCC early-stage melanomas and reduced epidermal integrity, thereby facilitating erosion of the epidermis and tumour ulceration.


Subject(s)
Melanoma , Skin Neoplasms , Transforming Growth Factor beta2/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Epidermis/metabolism , Humans , Melanoma/pathology , Skin Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Transforming Growth Factors/metabolism
2.
Clin Exp Dermatol ; 46(1): 109-117, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32687652

ABSTRACT

BACKGROUND: Chronic wounds continue to be a burden to healthcare systems, with ageing linked to increased prevalence of chronic wound development. Nutraceutical collagen peptides have been shown to reduce signs of skin ageing, but their therapeutic potential for cutaneous wound healing remains undefined. AIM: To determine the potential for nutraceutical collagen peptides to promote cutaneous wound healing in vitro in the context of age. METHODS: The potential for bovine- or porcine-derived nutraceutical collagen peptides to promote wound healing of primary cutaneous fibroblasts and keratinocytes derived from young and aged individuals in vitro was assessed by two-dimensional scratch and cell-viability assays and by immunofluorescence for the cell proliferation marker, Ki67. The achievement of peptide concentrations in vivo, equivalent to those exerting a beneficial effect on wound healing in vitro, was confirmed by pharmacokinetic studies of hydroxyproline, a biomarker for collagen peptide absorption, following peptide ingestion by healthy individuals over a wide age range. RESULTS: Results demonstrated significant nutraceutical collagen peptide-induced wound closure of both young and aged fibroblasts and keratinocytes, mediated by enhanced cellular proliferation and migration. Analysis of blood levels of hydroxyproline in young and aged individuals following porcine collagen peptide ingestion revealed peak serum/plasma levels after 2 h at similar concentrations to those exerting beneficial effects on wound healing in vitro. CONCLUSION: These data demonstrate the capacity for nutraceutical collagen peptides to promote cutaneous wound closure in vitro, at pharmacologically achievable concentrations in vivo, thereby offering a potential novel therapeutic strategy for the management of cutaneous wounds in young and aged individuals.


Subject(s)
Collagen/pharmacology , Dietary Supplements , Skin/drug effects , Wound Healing/drug effects , Adolescent , Adult , Aged , Animals , Blotting, Western , Cattle , Cell Proliferation , Fibroblasts/physiology , Humans , In Vitro Techniques , Keratinocytes/physiology , Male , Middle Aged , Skin Aging , Skin Physiological Phenomena , Swine , Young Adult
3.
Br J Dermatol ; 182(1): 156-165, 2020 01.
Article in English | MEDLINE | ID: mdl-31056744

ABSTRACT

BACKGROUND: The updated American Joint Committee on Cancer (AJCC) staging criteria for melanoma remain unable to identify high-risk stage I tumour subsets. OBJECTIVES: To determine the utility of epidermal autophagy and beclin 1 regulator 1 (AMBRA1)/loricrin (AMLo) expression as a prognostic biomarker for AJCC stage I cutaneous melanoma. METHODS: Peritumoral AMBRA1 expression was evaluated in a retrospective discovery cohort of 76 AJCC stage I melanomas. AMLo expression was correlated with clinical outcomes up to 12 years in two independent powered, retrospective validation and qualification cohorts comprising 379 AJCC stage I melanomas. RESULTS: Decreased AMBRA1 expression in the epidermis overlying primary melanomas in a discovery cohort of 76 AJCC stage I tumours was associated with a 7-year disease-free survival (DFS) rate of 81·5% vs. 100% survival with maintained AMBRA1 (P < 0·081). Following an immunohistochemistry protocol for semi-quantitative analysis of AMLo, analysis was undertaken in validation (n = 218) and qualification cohorts (n = 161) of AJCC stage I melanomas. Combined cohort analysis revealed a DFS rate of 98·3% in the AMLo low-risk group (n = 239) vs. 85·4% in the AMLo high-risk cohort (n = 140; P < 0·001). Subcohort multivariate analysis revealed that an AMLo hazard ratio (HR) of 4·04 [95% confidence interval (CI) 1·69-9·66; P = 0·002] is a stronger predictor of DFS than Breslow depth (HR 2·97, 95% CI 0·93-9·56; P = 0·068) in stage IB patients. CONCLUSIONS: Loss of AMLo expression in the epidermis overlying primary AJCC stage I melanomas identifies high-risk tumour subsets independently of Breslow depth. What's already known about this topic? There is an unmet clinical need for biomarkers of early-stage melanoma. Autophagy and beclin 1 regulator 1 (AMBRA1) is a proautophagy regulatory protein with known roles in cell proliferation and differentiation, and is a known tumour suppressor. Loricrin is a marker of epidermal terminal differentiation. What does this study add? AMBRA1 has a functional role in keratinocyte/epidermal proliferation and differentiation. The combined decrease/loss of peritumoral AMBRA1 and loricrin is associated with a significantly increased risk of metastatic spread in American Joint Committee on Cancer (AJCC) stage I tumours vs. melanomas, in which peritumoral AMBRA1 and loricrin are maintained, independently of Breslow depth. What is the translational message? The integration of peritumoral epidermal AMBRA1/loricrin biomarker expression into melanoma care guidelines will facilitate more accurate, personalized risk stratification for patients with AJCC stage I melanomas, thereby facilitating stratification for appropriate follow-up and informing postdiagnostic investigations, including sentinel lymph node biopsy, ultimately resulting in improved disease outcomes and rationalization of healthcare costs.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Melanoma , Membrane Proteins/genetics , Skin Neoplasms , Autophagy , Epidermis/pathology , Humans , Melanoma/pathology , Neoplasm Staging , Prognosis , Retrospective Studies , Skin Neoplasms/pathology , United States
4.
Br J Dermatol ; 180(2): 346-356, 2019 02.
Article in English | MEDLINE | ID: mdl-30339727

ABSTRACT

BACKGROUND: Patients with malignant melanoma often relapse after treatment with BRAF and/or mitogen-activated protein kinase kinase (MEK) inhibitors (MEKi) owing to development of drug resistance. OBJECTIVES: To establish the temporal pattern of CD271 regulation during development of resistance by melanoma to trametinib, and determine the association between development of resistance to trametinib and induction of prosurvival autophagy. METHODS: Immunohistochemistry for CD271 and p62 was performed on human naevi and primary malignant melanoma tumours. Western blotting was used to analyse expression of CD271, p62 and LC3 in melanoma subpopulations. Flow cytometry and immunofluorescence microscopy was used to evaluate trametinib-induced cell death and CD271 expression. MTS viability assays and zebrafish xenografts were used to evaluate the effect of CD271 and autophagy modulation on trametinib-resistant melanoma cell survival and invasion, respectively. RESULTS: CD271 and autophagic signalling are increased in stage III primary melanomas vs. benign naevi. In vitro studies demonstrate MEKi of BRAF-mutant melanoma induced cytotoxic autophagy, followed by the emergence of CD271-expressing subpopulations. Trametinib-induced CD271 reduced autophagic flux, leading to activation of prosurvival autophagy and development of MEKi resistance. Treatment of CD271-expressing melanoma subpopulations with RNA interference and small-molecule inhibitors to CD271 reduced the development of MEKi resistance, while clinically applicable autophagy modulatory agents - including Δ9-tetrahydrocannabinol and Vps34 - reduced survival of MEKi-resistant melanoma cells. Combined MEK/autophagy inhibition also reduced the invasive and metastatic potential of MEKi-resistant cells in an in vivo zebrafish xenograft. CONCLUSIONS: These results highlight a novel mechanism of MEKi-induced drug resistance and suggest that targeting autophagy may be a translatable approach to resensitize drug-resistant melanoma cells to the cytotoxic effects of MEKi.


Subject(s)
Autophagy/drug effects , Drug Resistance, Neoplasm/immunology , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Skin Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/immunology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Melanoma/immunology , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Nevus/immunology , Nevus/pathology , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism , Receptors, Nerve Growth Factor/analysis , Receptors, Nerve Growth Factor/metabolism , Skin/immunology , Skin/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays , Zebrafish
5.
Br J Dermatol ; 175(6): 1210-1220, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27167239

ABSTRACT

BACKGROUND: Expression of the chemokine receptor CXCR4 is known to regulate melanoma metastasis to distant sites with high expression of the CXCL12 ligand. However, the prognostic impact of CXCR4 expression and potential for autocrine-mediated activation of prosurvival mitogen-activated protein kinase signalling remains enigmatic. Furthermore, expression of the decoy receptor CXCR7 within the local cutaneous melanoma microenvironment remains undefined. OBJECTIVES: To define the contribution and prognostic impact of CXCR4-CXCR7-CXCL12 signalling in primary cutaneous melanomas and the immediate tumour microenvironment. METHODS: Immunohistochemical/immunofluorescent expression of CXCR4, CXCR7 or CXC12 was analysed in human metastatic melanoma cell lines, primary cutaneous cell types and a retrospective cohort of primary melanomas/benign naevi. CXCL12 secretion by melanoma/cutaneous cells was evaluated by enzyme-linked immunosorbent assay, and autocrine CXCR4-CXCL12 signalling was investigated by addition of a CXCL12-neutralizing antibody. RESULTS: CXCR4 expression was significantly higher in primary melanomas that subsequently metastasized after 7 years (P = 0·037). Stratification for American Joint Committee on Cancer (AJCC) stage II disease revealed significantly decreased disease-free survival in patients with > 50% CXCR4 expression (P = 0·036), while comparative analysis of CXCL12 expression in the adjacent epidermis of all AJCC stage melanomas revealed increased CXCL12 correlated with prolonged time to metastasis (P = 0·014). CXCR7 was expressed within the primary melanoma microenvironment but was absent on primary tumours. Addition of anti-CXCL12 to BRAF-mutant melanoma cells resulted in downregulation of phospho-CXCR4 and phospho-extracellular signal-related kinase, indicating autocrine CXCR4-CXCL12 signalling. CONCLUSIONS: CXCR4 expression defines a potential prognostic biomarker for AJCC stage II melanoma. Moreover, targeting the CXCR4-CXCR7-CXCL12 axis may represent a novel therapeutic strategy to prevent early melanoma progression.


Subject(s)
Biomarkers, Tumor/metabolism , Chemokine CXCL12/metabolism , Melanoma/mortality , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Skin Neoplasms/mortality , Cell Line, Tumor , Down-Regulation/physiology , GTP Phosphohydrolases/genetics , Humans , Melanoma/metabolism , Membrane Proteins/genetics , Neoplasm Metastasis , Phenotype , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Retrospective Studies , Skin Neoplasms/metabolism , Tumor Microenvironment
6.
Cell Death Differ ; 22(6): 946-58, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25361077

ABSTRACT

The notorious unresponsiveness of metastatic cutaneous melanoma to current treatment strategies coupled with its increasing incidence constitutes a serious worldwide clinical problem. Moreover, despite recent advances in targeted therapies for patients with BRAF(V600E) mutant melanomas, acquired resistance remains a limiting factor and hence emphasises the acute need for comprehensive pre-clinical studies to increase the biological understanding of such tumours in order to develop novel effective and longlasting therapeutic strategies. Autophagy and ER stress both have a role in melanoma development/progression and chemoresistance although their real impact is still unclear. Here, we show that BRAF(V600E) induces a chronic ER stress status directly increasing basal cell autophagy. BRAF(V600E)-mediated p38 activation stimulates both the IRE1/ASK1/JNK and TRB3 pathways. Bcl-XL/Bcl-2 phosphorylation by active JNK releases Beclin1 whereas TRB3 inhibits the Akt/mTor axes, together resulting in an increase in basal autophagy. Furthermore, we demonstrate chemical chaperones relieve the BRAF(V600E)-mediated chronic ER stress status, consequently reducing basal autophagic activity and increasing the sensitivity of melanoma cells to apoptosis. Taken together, these results suggest enhanced basal autophagy, typically observed in BRAF(V600E) melanomas, is a consequence of a chronic ER stress status, which ultimately results in the chemoresistance of such tumours. Targeted therapies that attenuate ER stress may therefore represent a novel and more effective therapeutic strategy for BRAF mutant melanoma.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Apoptosis/genetics , Apoptosis/physiology , Autophagy/genetics , Autophagy/physiology , Cell Line, Tumor , Endoplasmic Reticulum Stress/genetics , Humans , Lentivirus/genetics , Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms , Melanoma, Cutaneous Malignant
7.
Br J Cancer ; 109(2): 433-43, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23807168

ABSTRACT

BACKGROUND: Glucose regulated protein 78 (GRP78) functions as a sensor of endoplasmic reticulum (ER) stress. The aim of this study was to test the hypothesis that molecules that bind to GRP78 induce the unfolded protein response (UPR) and enhance cell death in combination with ER stress inducers. METHODS: Differential scanning calorimetry (DSC), measurement of cell death by flow cytometry and the induction of ER stress markers using western blotting. RESULTS: Epigallocatechin gallate (EGCG), a flavonoid component of Green Tea Camellia sinensis, and honokiol (HNK), a Magnolia grandiflora derivative, bind to unfolded conformations of the GRP78 ATPase domain. Epigallocatechin gallate and HNK induced death in six neuroectodermal tumour cell lines tested. Levels of death to HNK were twice that for EGCG; half-maximal effective doses were similar but EGCG sensitivity varied more widely between cell types. Honokiol induced ER stress and UPR as predicted from its ability to interact with GRP78, but EGCG was less effective. With respect to cell death, HNK had synergistic effects on melanoma and glioblastoma cells with the ER stress inducers fenretinide or bortezomib, but only additive (fenretinide) or inhibitory (bortezomib) effects on neuroblastoma cells. CONCLUSION: Honokiol induces apoptosis due to ER stress from an interaction with GRP78. The data are consistent with DSC results that suggest that HNK binds to GRP78 more effectively than EGCG. Therefore, HNK may warrant development as an antitumour drug.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Biphenyl Compounds/therapeutic use , Catechin/analogs & derivatives , Heat-Shock Proteins/metabolism , Lignans/therapeutic use , Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/metabolism , Biphenyl Compounds/metabolism , Catechin/metabolism , Catechin/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Heat-Shock Proteins/antagonists & inhibitors , Humans , Lignans/metabolism , Molecular Targeted Therapy , Molecular Weight , Neoplasms/pathology , Protein Binding/drug effects
8.
Clin Exp Dermatol ; 38(4): 421-3, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23530461

ABSTRACT

Early-stage cutaneous squamous cell carcinoma (cSCC) has a favourable prognosis. Metastatic disease is probably associated with chemoresistance mediated through the activation of pro-survival phosphatidylinositol 3-kinase/AKT signalling. Inhibition of activated AKT partially increases chemosensitivity but induces autophagy, the principal lysosomal mechanism for the bulk degradation and recycling of proteins and damaged organelles. The aim of the current study was to test the hypothesis that combined inhibition of AKT signalling and autophagy by the lysosomal inhibitor chloroquine increases the susceptibility to docetaxel-induced apoptosis of cSCC cells isolated from a lymph-node metastasis. Combined AKT inhibition and chloroquine treatment of MET 4 cSCC cells resulted in significantly enhanced inhibition of cell viability and apoptosis induced by clinically achievable concentrations of docetaxel (P < 0.001). Inhibition of both autophagy and AKT thus represents an effective and viable therapeutic strategy to increase the cytotoxicity of docetaxel for the treatment of advanced cSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Squamous Cell/drug therapy , Enzyme Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Skin Neoplasms/drug therapy , Taxoids/pharmacology , Analysis of Variance , Antimalarials/pharmacology , Carcinoma, Squamous Cell/enzymology , Chloroquine/pharmacology , Docetaxel , Dose-Response Relationship, Drug , Drug Therapy, Combination , Humans , Signal Transduction/drug effects , Skin Neoplasms/enzymology , Tumor Cells, Cultured
9.
Br J Cancer ; 108(8): 1634-40, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23538388

ABSTRACT

BACKGROUND: Despite intensive research and novel adjuvant therapies, there is currently no cure for metastatic melanoma. The chemokine receptor CXCR4 controls metastasis to sites such as the liver; however, the therapeutic blockade with the existing agents has proven difficult. METHODS: AMD11070, a novel orally bioavailable inhibitor of CXCR4, was tested for its ability to inhibit the migration of melanoma cells compared with the commonly described antagonist AMD3100. RESULTS: AMD11070 abrogated melanoma cell migration and was significantly more effective than AMD3100. Importantly for the clinical context, the expression of B-RAF-V600E did not the affect the sensitivity of AMD11070. CONCLUSION: Liver-resident myofibroblasts excrete CXCL12, which is able to promote the migration of CXCR4-expressing tumour cells from the blood into the liver. Blockade of this axis by AMD11070 thus represents a novel therapeutic strategy for both B-RAF wild-type and mutated melanomas.


Subject(s)
Aminoquinolines/pharmacology , Benzimidazoles/pharmacology , Cell Migration Inhibition/drug effects , Chemokine CXCL12/antagonists & inhibitors , Melanoma/drug therapy , Melanoma/pathology , Receptors, CXCR4/antagonists & inhibitors , Butylamines , Cell Line, Tumor , Flow Cytometry , Heterocyclic Compounds, 1-Ring , Humans , Liver Neoplasms/secondary
10.
Br J Cancer ; 101(8): 1448-55, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19724275

ABSTRACT

BACKGROUND: Metastatic melanoma is the most deadly form of skin cancer and with an overall 5-year survival rate of <11%, there is an acute need for novel therapeutic strategies. Activating mutations in the BRAF oncogene are present in 50-70% of cases and contribute to tumourigenesis, thus, defining downstream targets of oncogenic BRAF may help define novel targets for therapeutic intervention. The Ca(2+)/calcineurin-regulated transcription factor, Nuclear factor of activated T-cells (NFAT), is important in the pathogenesis of several human cancers, target genes of which are also known to contribute to melanoma progression. One such NFAT target gene is COX-2, increased expression of which correlates with poor prognosis; however, upstream regulators of COX-2 in melanoma remain undefined. Therefore, the aim of this study was to evaluate NFAT expression and activity in metastatic melanoma and establish whether or not oncogenic BRAF signalling modulates NFAT activity and determine if NFAT is a key upstream regulator of COX-2 in melanoma. METHODS: Nuclear factor of activated T-cells transcriptional activity and protein expression were determined in three human metastatic melanoma cell lines with differing B-RAF mutational status. NFAT activation by oncogenic BRAF(V600E) was explored by BRAF(V600E) overexpression and application of the specific MEK inhibitor PD98059. Regulation of COX-2 expression by NFAT was investigated using NFAT-targeted siRNA, calcineurin inhibitors cyclosporin A and FK506, in addition to COX-2 luciferase reporter vectors that selectively lacked NFAT binding sites. RESULTS: NFAT transcriptional activity was increased in BRAF-mutated melanoma cells compared with wild-type cells. Furthermore, in wild-type cells, overexpression of BRAF(V600E) increased NFAT activity, which was blocked by the MEK inhibitor PD98059. Using calcineurin inhibitors and siRNA-mediated knockdown of NFAT2 and 4, we show NFAT is required for COX-2 promoter activation and protein induction in metastatic melanoma cells. CONCLUSION: NFAT2 and 4 are expressed in human metastatic melanoma cell lines and are activated by oncogenic BRAF(V600E) via MEK/ERK signalling. NFAT is an important upstream regulator of COX-2 in metastatic melanoma. Furthermore, as the BRAF/MEK/ERK pathway is hyperactive in other malignancies and MEK/ERK are also activated by oncogenic RAS in 30% of all human cancers, the potential to exploit NFAT signalling for therapeutic benefit warrants further investigation.


Subject(s)
Melanoma/secondary , NFATC Transcription Factors/physiology , Proto-Oncogene Proteins B-raf/physiology , Calcineurin Inhibitors , Cell Line, Tumor , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Humans , Melanoma/therapy , NFATC Transcription Factors/antagonists & inhibitors , Promoter Regions, Genetic , Signal Transduction
11.
J Clin Pathol ; 61(10): 1098-103, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18682419

ABSTRACT

BACKGROUND: MYCN is the most commonly amplified gene in human neuroblastomas. This proto-oncogene has been overexpressed in a mouse model of the disease in order to explore the role of MYCN in this tumour. AIMS: To report the histopathological features of neuroblastomas from MYCN transgenic mice. METHODS: 27 neuroblastomas from hemizygous transgenic mice and four tumours from homozygous mice were examined histologically; Ki67 and MYCN immunocytochemistry was performed in 24 tumours. RESULTS: Tumours obtained from MYCN transgenic mice resembled human neuroblastomas, displaying many of the features associated with stroma-poor neuroblastoma, including heterogeneity of differentiation (but no overt ganglionic differentiation was seen), low levels of Schwannian stroma and a high mitosis karyorrhexis index. The tumours had a median Ki67 labelling index of 70%; all tumours expressed MYCN with a median labelling index of 68%. The most striking difference between the murine and human neuroblastomas was the presence of tingible body macrophages in the transgenic mouse tumours reflecting high levels of apoptosis. This has not previously been described in human or other murine neuroblastoma models. CONCLUSIONS: These studies highlight the histological similarities between tumours from MYCN transgenic mice and human neuroblastomas, and reaffirm their role as a valuable model to study the biology of aggressive human neuroblastoma.


Subject(s)
Abdominal Neoplasms/pathology , Neuroblastoma/pathology , Nuclear Proteins , Oncogene Proteins , Abdominal Neoplasms/genetics , Animals , Biomarkers/analysis , Blotting, Western , Female , Gene Amplification , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Male , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Oncogene Proteins/analysis , Oncogene Proteins/genetics , Proto-Oncogene Mas , Ubiquitin Thiolesterase/analysis
12.
Br J Cancer ; 96(7): 1062-71, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17353921

ABSTRACT

Endoplasmic reticulum (ER) malfunction, leading to ER stress, can be a consequence of genome instability and hypoxic tissue environments. Cancer cells survive by acquiring or enhancing survival mechanisms to counter the effects of ER stress and these homeostatic responses may be new therapeutic targets. Understanding the links between ER stress and apoptosis may be approached using drugs specifically to target ER stress responses in cancer cells. The retinoid analogue fenretinide [N-(4-hydroxyphenyl) retinamide] is a new cancer preventive and chemotherapeutic drug, that induces apoptosis of some cancer cell types via oxidative stress, accompanied by induction of an ER stress-related transcription factor, GADD153. The aim of this study was to test the hypothesis that fenretinide induces ER stress in neuroectodermal tumour cells, and to elucidate the role of ER stress responses in fenretinide-induced apoptosis. The ER stress genes ERdj5, ERp57, GRP78, calreticulin and calnexin were induced in neuroectodermal tumour cells by fenretinide. In contrast to the apoptosis-inducing chemotherapeutic drugs vincristine and temozolomide, fenretinide induced the phosphorylation of eIF2alpha, expression of ATF4 and splicing of XBP-1 mRNA, events that define ER stress. In these respects, fenretinide displayed properties similar to the ER stress inducer thapsigargin. ER stress responses were inhibited by antioxidant treatment. Knockdown of ERp57 or ERdj5 by RNA interference in these cells increased the apoptotic response to fenretinide. These data suggest that downregulating homeostatic ER stress responses may enhance apoptosis induced by oxidative stress-inducing drugs acting through the ER stress pathway. Therefore, ER-resident proteins such as ERdj5 and ERp57 may represent novel chemotherapeutic targets.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum/metabolism , Fenretinide/pharmacology , Molecular Chaperones/metabolism , Oxidative Stress , Protein Disulfide-Isomerases/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Alternative Splicing , Biomarkers, Tumor/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , HSP40 Heat-Shock Proteins , Humans , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroectodermal Tumors/drug therapy , Neuroectodermal Tumors/metabolism , Neuroectodermal Tumors/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Regulatory Factor X Transcription Factors , Transcription Factors , Tumor Cells, Cultured/drug effects , X-Box Binding Protein 1
13.
Apoptosis ; 10(3): 493-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15909111

ABSTRACT

Fenretinide induces apoptosis in SH-SY5Y neuroblastoma cells via a signaling pathway involving the production of reactive oxygen species (ROS), 12-lipoxygenase activity and the induction of the GADD153 transcription factor. NF-kappa B is a key element of many cell signaling pathways and adopts a pro- or anti-apoptotic role in different cell types. Studies have suggested that NF-kappa B may play a pro-apoptotic role in SH-SY5Y cells, and in other cell types NF-kappa B activation may be linked to lipoxygenase activity. The aim of this study was to test the hypothesis that NF-kappa B activity mediates fenretinide-induced apoptosis in SH-SY5Y neuroblastoma cells. Using a dominant-negative construct for Ikappa Balpha stably transfected into SH-SY5Y cells, we show that apoptosis, but not the induction of ROS, in response to fenretinide was blocked by abrogation of NF-kappa B activity. In parental SH-SY5Y cells, fenretinide induced NF-kappa B activity and Ikappa Balpha phosphorylation. These results suggest that NF-kappa B activity links fenretinide-induced ROS to the induction of apoptosis in SH-SH5Y cells, and may be a target for the future development of drugs for neuroblastoma therapy.


Subject(s)
Apoptosis/drug effects , Fenretinide/pharmacology , NF-kappa B/physiology , Flow Cytometry , Humans , I-kappa B Proteins/biosynthesis , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , Neuroblastoma , Phosphorylation , Reactive Oxygen Species/metabolism , Transfection , Tumor Cells, Cultured
14.
Med Pediatr Oncol ; 36(1): 115-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11464861

ABSTRACT

BACKGROUND AND PROCEDURE: The CD95/CD95 ligand (CD95L) system is a key regulator of apoptosis. To evaluate a possible role of the CD95/CD95L system in human neuroblastoma (NB) cells, we investigated the constitutive and interferongamma (INFgamma)-induced expression of CD95 and CD95L, and CD95-mediated cell death in the SK-N-BE(2) cell line. RESULTS: Modulation of CD95/CD95L expression and triggering of an autocrine apoptotic mechanism by IFNgamma suggest a potential role for INFgamma as a therapeutic agent for NB. CONCLUSIONS: The evidence that retinoids induce apoptosis via tissue transglutaminase (tTG) and that N-methyl-D-aspartate (NMDA) and gp120 act through the nitric oxide synthase (NOS) activation pathway, indicates the existence of different molecular mechanisms of action, whose pharmacological exploitation might be used in an additive fashion.


Subject(s)
Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Interferon-gamma/pharmacology , Membrane Glycoproteins/physiology , Neoplasm Proteins/physiology , Neuroblastoma/pathology , fas Receptor/physiology , Calcium/physiology , Calcium Channel Blockers/pharmacology , Drug Synergism , Excitatory Amino Acid Antagonists/pharmacology , Fas Ligand Protein , GTP-Binding Proteins/physiology , HIV Envelope Protein gp120/physiology , Humans , Ion Transport , Membrane Glycoproteins/genetics , N-Methylaspartate/pharmacology , Neoplasm Proteins/genetics , Neuroblastoma/metabolism , Nitric Oxide/physiology , Nitric Oxide Synthase/physiology , Protein Glutamine gamma Glutamyltransferase 2 , Receptors, N-Methyl-D-Aspartate/drug effects , Transglutaminases/physiology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology , fas Receptor/genetics
15.
Med Pediatr Oncol ; 36(1): 135-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11464866

ABSTRACT

BACKGROUND: 9-cis retinoic acid (RA) is more effective than all-trans RA at inducing neuroblastoma differentiation in vitro, and has distinct biological properties with respect to its ability to promote apoptosis in N-type neuroblastoma cells. The cellular effects of 9-cis RA may, in part, result from activation of retinoid X receptor (RXR) homodimers. If this hypothesis is correct, 9-cis RA may control the expression of a different subset of retinoid-regulated genes compared to all-trans RA. PROCEDURE: We have therefore used differential mRNA display to identify genes differentially expressed in neuroblastoma cells in response to all-trans and 9-cis RA. RESULTS: The majority of cDNAs differentially expressed in response to all-trans or 9-cis RA matched to nonredundant Genbank sequences or EST database sequences. Differential-display profiles were similar in SH SY 5Y and SH S EP cells, clonal derivatives of the mixed neuroblastoma cell line SK N SH, although there were apparent differences between these cell lines with respect to the retinoid-regulation of specific RT-PCR cDNA fragments. CONCLUSIONS: These data support the view that 9-cis and all-trans RA act via different receptor mechanisms.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Tretinoin/pharmacology , Alitretinoin , Apoptosis/drug effects , DNA, Complementary/genetics , Expressed Sequence Tags , Humans , Neoplasm Proteins/drug effects , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Transcription, Genetic/drug effects , Tumor Cells, Cultured/drug effects
16.
Med Pediatr Oncol ; 35(6): 663-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11107142

ABSTRACT

The RARbeta/gamma-selective retinoids fenretinide and CD437 induce caspase-dependent apoptosis but generate free radicals independently of caspases. Apoptosis, but not free radical generation, induced by these retinoids is inhibited by RARbeta/gamma-specific antagonists. Both fenretinide and CD437 induce apoptosis synergistically with cisplatin, carboplatin, or etoposide. However, antioxidants inhibit this synergy to the level obtained with chemotherapeutic drugs alone, and this implies that free radical generation is important in the synergistic response. Since apoptosis induced by fenretinide or CD437 is mediated by apoptotic pathways involving RARs and/or mitochondria and differs from mechanisms of chemotherapy-induced apoptosis this may explain the strong synergistic response seen between these synthetic retinoids and chemotherapeutic drugs. These results suggest that fenretinide or CD437 may be useful adjuncts to neuroblastoma therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Fenretinide/therapeutic use , Neuroblastoma/drug therapy , Retinoids/therapeutic use , Apoptosis/drug effects , Child , Drug Synergism , Free Radicals/metabolism , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , Tumor Cells, Cultured
17.
Int J Cancer ; 88(6): 977-85, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11093824

ABSTRACT

Retinoic acid therapy improves the survival of children with neuroblastoma and 13-cis retinoic acid now forms an important component of treatment for residual disease of stage IV neuroblastoma after chemotherapy. However, although 13-cis retinoic acid induces differentiation, other retinoids are effective at inducing apoptosis of neuroblastoma in vitro, including the novel compounds fenretinide and CD437 and these may be alternative retinoids for neuroblastoma therapy. The aim of our study was to evaluate the ability of fenretinide, CD437 (6-¿3-(1-adamantyl)-4-hydroxyphenyl¿ -2-naphthalene carboxylic acid) and different retinoic acid isomers to induce apoptosis of neuroblastoma in conjunction with the chemotherapeutic drugs, cisplatin, etoposide and carboplatin. Neuroblastoma cell lines were treated with retinoids prior to treatment with chemotherapeutic agents and flow cytometry used to measure apoptosis and free radical generation. Pre-treatment of neuroblastoma cell lines with fenretinide or CD437 prior to treatment with cisplatin, etoposide or carboplatin synergistically increased apoptosis, an effect not seen with 13-cis, all-trans or 9-cis retinoic acid. Contrary to retinoic acid isomers or chemotherapeutic drugs, apoptosis of neuroblastoma cells induced by fenretinide or CD437 was accompanied by the generation of intracellular free radicals. Quenching of fenretinide- or CD437-induced free radicals with antioxidants abolished the synergistic response seen with the subsequent addition of chemotherapeutic agents. Therefore, the generation of free radicals by fenretinide or CD437 may be the key property of these retinoids leading to synergistic responses with chemotherapeutic drugs. Clearly, these synthetic retinoids provide new opportunities for novel neuroblastoma therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Neuroblastoma/drug therapy , Carboplatin/therapeutic use , Cell Survival , Cisplatin/therapeutic use , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Drug Synergism , Etoposide/therapeutic use , Fenretinide/administration & dosage , Flow Cytometry , Free Radicals/analysis , Humans , Neuroblastoma/physiopathology , Retinoids/administration & dosage , Time Factors , Tretinoin/therapeutic use , Tumor Cells, Cultured/drug effects
18.
Exp Cell Res ; 260(1): 50-60, 2000 Oct 10.
Article in English | MEDLINE | ID: mdl-11010810

ABSTRACT

Fenretinide is an effective inducer of apoptosis in many malignancies but its precise mechanism(s) of action in the induction of apoptosis in neuroblastoma is unclear. To characterize fenretinide-induced apoptosis, neuroblastoma cell lines were treated with fenretinide and flow cytometry was used to measure apoptosis, free radical generation, and mitochondrial permeability changes. Fenretinide induced high levels of caspase-dependent apoptosis accompanied by an increase in free radicals and the release of cytochrome c in the absence of mitochondrial permeability transition. Apoptosis was blocked by two retinoic acid receptor (RAR)-beta/gamma-specific antagonists, but not by an RARalpha-specific antagonist. Free radical induction in response to fenretinide was not blocked by the caspase inhibitor ZVAD or by RAR antagonists and was only marginally reduced in cells selected for resistance to fenretinide. Therefore, free radical generation may be only one of a number of intracellular mechanisms of apoptotic signaling in response to fenretinide. These results suggest that the effector pathway of fenretinide-induced apoptosis of neuroblastoma is caspase dependent, involving mitochondrial release of cytochrome c independently of permeability changes, and mediated by specific RARs. As the mechanism of action of fenretinide may be different from other retinoids, this compound may be a valuable adjunct to neuroblastoma therapy with retinoic acid and conventional chemotherapeutic drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Fenretinide/pharmacology , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/physiology , Caspase Inhibitors , Caspases/metabolism , Cell Membrane Permeability/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Cytochrome c Group/metabolism , Free Radicals/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma/metabolism , Oligopeptides/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Receptors, Retinoic Acid/antagonists & inhibitors , Tumor Cells, Cultured
19.
FEBS Lett ; 445(2-3): 415-9, 1999 Feb 26.
Article in English | MEDLINE | ID: mdl-10094499

ABSTRACT

Retinoic acid modulates growth and induces differentiation and apoptosis of neuroblastoma cells in vitro, with the all-trans and 9-cis isomers having different biological properties. Transcriptional activation in response to retinoic acid isomers is mediated by retinoic acid receptors and retinoid X receptors. The differential expression of co-activators and co-repressors which preferentially interact with retinoic acid receptors or retinoid X receptors may be a mechanism leading to different cellular responses to 9-cis and all-trans retinoic acid. To test this hypothesis, we have studied the expression of the nuclear receptor co-regulators TIF1alpha, TIF1beta, SUG1 and SMRT in the N-type and S-type neuroblastoma cell lines SH SY 5Y and SH S EP. Transcripts for all four co-regulators were expressed in these neuroblastoma cells. The expression of TIF1alpha, TIF1beta and SUG1 did not change in response to retinoic acid; however, SMRT was induced in both neuroblastoma cell lines, but particularly by all-trans retinoic acid in SH S EP cells. An additional co-activator, Trip3, was isolated by differential mRNA display and shown to be preferentially induced by 9-cis retinoic acid in SH SY 5Y and SH S EP cells. These data suggest that retinoic acid isomer-specific induction of nuclear receptor co-regulators may determine, in part, the differential biological effects of retinoic acid isomers.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Nuclear Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Tretinoin/metabolism , ATPases Associated with Diverse Cellular Activities , Humans , Intracellular Signaling Peptides and Proteins , Isomerism , LIM Domain Proteins , Neuroblastoma , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 2 , Proteasome Endopeptidase Complex , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Transcription Factors/metabolism , Tretinoin/pharmacology , Tripartite Motif-Containing Protein 28 , Tumor Cells, Cultured
20.
Eur J Cancer ; 34(1): 111-7, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9624247

ABSTRACT

The aim of this study was to investigate in vitro the effects of all-trans retinoic acid (RA), 9-cis RA and the RXR-selective analogue, LG69, on the morphological differentiation, proliferation and gene expression of neuroblastoma cells. Three different cell lines were cultured with the retinoid for either 9 continuous days or for 5 days followed by 4 days without the retinoid and morphological differentiation was assessed both qualitatively and quantitatively. SH SY 5Y cell proliferation was examined by measuring cell numbers after exposure to the retinoids and RAR-beta gene expression was examined by Northern blot analysis. Morphological differentiation was more effectively induced by all-trans and 9-cis RA than by LG69. SH SY 5Y cells, when treated with 9-cis RA for only 5 of the 9 days of culture, underwent apoptosis, but this was not seen with 9 days continuous exposure nor with LG69. Inhibition of SH SY 5Y cell proliferation by all-trans or 9-cis RA was dose-dependent, but LG69 had little effect. Conversely, LG69 induced higher expression of RAR-beta than all-trans RA, but less than that produced by 9-cis RA. These data suggest that 9-cis RA as a single agent is the most effective modulator of neuroblastoma behaviour and may be the most appropriate therapeutic agent.


Subject(s)
Antineoplastic Agents/pharmacology , Neuroblastoma/pathology , Tetrahydronaphthalenes/pharmacology , Tretinoin/pharmacology , Bexarotene , Blotting, Northern , Cell Differentiation/drug effects , Cell Division/drug effects , Dose-Response Relationship, Drug , Gene Expression , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...