Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Tumour Biol ; 43(1): 11-26, 2021.
Article En | MEDLINE | ID: mdl-33935126

BACKGROUND: Retinoblastoma (RB) is the most common childhood eye cancer. Chemotherapeutic drugs such as etoposide used in RB treatment often cause massive side effects and acquired drug resistances. Dysregulated genes and miRNAs have a large impact on cancer progression and development of chemotherapy resistances. OBJECTIVE: This study was designed to investigate the involvement of retinoic acid receptor alpha (RARα) in RB progression and chemoresistance as well as the impact of miR-138, a potential RARα regulating miRNA. METHODS: RARα and miR-138 expression in etoposide resistant RB cell lines and chemotherapy treated patient tumors compared to non-treated tumors was revealed by Real-Time PCR. Overexpression approaches were performed to analyze the effects of RARα on RB cell viability, apoptosis, proliferation and tumorigenesis. Besides, we addressed the effect of miR-138 overexpression on RB cell chemotherapy resistance. RESULTS: A binding between miR-138 and RARα was shown by dual luciferase reporter gene assay. The study presented revealed that RARα is downregulated in etoposide resistant RB cells, while miR-138 is endogenously upregulated. Opposing RARα and miR-138 expression levels were detectable in chemotherapy pre-treated compared to non-treated RB tumor specimen. Overexpression of RARα increases apoptosis levels and reduces tumor cell growth of aggressive etoposide resistant RB cells in vitro and in vivo. Overexpression of miR-138 in chemo-sensitive RB cell lines partly enhances cell viability after etoposide treatment. CONCLUSIONS: Our findings show that RARα acts as a tumor suppressor in retinoblastoma and is downregulated upon etoposide resistance in RB cells. Thus, RARα may contribute to the development and progression of RB chemo-resistance.


Drug Resistance, Neoplasm/drug effects , Etoposide/pharmacology , MicroRNAs/metabolism , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Retinoic Acid Receptor alpha/metabolism , 3' Untranslated Regions , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Drug Resistance, Neoplasm/genetics , Etoposide/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Protein Binding , Retinal Neoplasms/diet therapy , Retinal Neoplasms/genetics , Retinoblastoma/drug therapy , Retinoblastoma/genetics , Retinoic Acid Receptor alpha/genetics
2.
Int J Mol Sci ; 20(17)2019 Aug 24.
Article En | MEDLINE | ID: mdl-31450568

Trefoil factor family peptide 3 (TFF3) is supposed to have tumor suppressive functions in retinoblastoma (RB), but the functional pathway is not completely understood. In the study presented, we investigated the downstream pathway of TFF3 signaling in Y79 RB cells. Results from pG13-luciferase reporter assays and western blot analyses indicate induced p53 activity with an upregulation of miR-34a after TFF3 overexpression. Expression levels of the predicted miR-34a target epithelial membrane protein 1 (EMP1) are reduced after TFF3 overexpression. As revealed by WST-1 assay, BrdU, and DAPI cell counts viability and proliferation of Y79 cells significantly decrease following EMP1 knockdown, while apoptosis levels significantly increase. Opposite effects on Y79 cells' growth could be shown after EMP1 overexpression. Caspase assays showed that EMP1 induced apoptosis after overexpression is at least partially caspase-3/7 dependent. Colony formation and soft agarose assays, testing for anchorage independent growth, revealed that EMP1 overexpressing Y79 cells have a significantly higher ability to form colonies. In in ovo chicken chorioallantoic membrane (CAM) assays inoculated EMP1 overexpressing Y79 cells form significantly larger CAM tumors. Moreover, miR-34a overexpression increases sensitivity of Y79 cells towards RB chemotherapeutics, however, without involvement of EMP1. In summary, the TFF3 signaling pathway in Y79 RB cells involves the activation of p53 with downstream induction of miR-34a and subsequent inhibition of EMP1.


Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA Interference , Receptors, Cell Surface/genetics , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Knockdown Techniques , Genes, Reporter , Humans , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Retinoblastoma/genetics , Retinoblastoma/metabolism , Trefoil Factor-3
3.
Oncol Rep ; 39(1): 160-172, 2018 Jan.
Article En | MEDLINE | ID: mdl-29192327

Retinoblastoma (RB) is the most common malignant intraocular tumor in early childhood. Imminent chemotherapy resistance diminishes the clinical-therapeutic options and emphasizes the necessity for new therapeutic approaches. The present study aimed at characterizing and comparing etoposide and cisplatin-resistant human RB cell lines with regard to changes in proliferation and apoptosis levels, anchorage independent growth behavior in vitro as well as tumor formation capacity in vivo. The proliferation rates were significantly increased in the etoposide-resistant RB cell lines Y-79, WERI-Rb1 and RB-355 reflecting significantly higher growth kinetics compared to the parental controls. In line with these findings in in vivo chicken chorioallantoic (CAM) assays, etoposide-resistant cell lines generated significantly increased numbers of tumors with higher tumor weights compared to their parental counterparts. In contrast to etoposide, the cisplatin-resistant RB cell lines Y-79, WERI-Rb1 and RB-355 displayed significantly increased apoptosis rates and reduced proliferation rates resulting in significantly decreased growth kinetics. Tumor formation capacity of cisplatin-resistant cell lines did not significantly change, and in comparison with parental controls cisplatin-resistant Y-79 cells displayed significantly reduced tumor weight. Soft agarose assays indicated that anchorage-independent growth of all chemotherapy-resistant cell lines analyzed was significantly decreased. Summarizing, one can state that etoposide-resistant RB cells behave more aggressively than the tumor cells of origin and potentially represent a risk factor for local relapse, while cisplatin-resistant cells show a significantly decreased tumorigenic potential.


Cisplatin/pharmacology , Drug Resistance, Neoplasm , Etoposide/pharmacology , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chickens , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/pathology , Humans , Neoplasm Transplantation , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy
4.
Int J Cancer ; 141(3): 549-560, 2017 08 01.
Article En | MEDLINE | ID: mdl-28481041

Trefoil factor family (TFF) peptides have been shown to play a pivotal role in oncogenic transformation, tumorigenesis and metastasis by changing cell proliferation, apoptosis, migration and invasion behavior of various cancer cell lines. In the study presented, we investigated the effect of TFF1 overexpression on cell growth, viability, migration and tumorigenicity of different retinoblastoma (RB) cell lines. Transient TFF1 overexpression significantly increases RB cell apoptosis levels. Stable, lentiviral TFF1 overexpression likewise decreases RB cell viability, proliferation and growth and significantly increases apoptosis as revealed by WST-1 assays, BrdU and DAPI cell counts. TFF1-induced apoptosis is executed via cleaved caspase-3 activation as revealed by caspase blockage experiments and caspase-3 immunocytochemistry. Results from pG13-luciferase reporter assays and Western blot analyses indicate that TFF1-induced apoptosis is mediated through transcriptional activity of p53 with concurrently downregulated miR-18a expression. In ovo chicken chorioallantoic membrane (CAM) assays revealed that TFF1 overexpression significantly decreases the size of tumors forming from Y79 and RB355 cells and reduces the migration potential of RB355 cells. Differentially expressed genes and pathways involved in cancer progression were identified after TFF1 overexpression in Y79 cells by gene expression array analysis, underlining the effects on reduced tumorigenicity. TFF1 knockdown in RBL30 cells revealed caspase-3/7-independent apoptosis induction, but no changes on cell proliferation level. In summary, the in vitro and in vivo data demonstrate for the first time a tumor suppressor function of TFF1 in RB cells which is at least partly mediated by p53 activation and miR-18a downregulation.


Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma/pathology , Trefoil Factor-1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Caspases/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chick Embryo , Chickens , Chorioallantoic Membrane , Humans , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retinoblastoma Protein/genetics , Trefoil Factor-1/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
5.
PLoS One ; 11(9): e0163025, 2016.
Article En | MEDLINE | ID: mdl-27626280

Trefoil factor family (TFF) peptides have been shown to effect cell proliferation, apoptosis, migration and invasion of normal cells and various cancer cell lines. In the literature TFF peptides are controversially discussed as tumor suppressors and potential tumor progression factors. In the study presented, we investigated the effect of TFF3 overexpression on growth, viability, migration and tumorigenicity of the human retinoblastoma cell lines Y-79, WERI-Rb1, RBL-13 and RBL-15. As revealed by WST-1 and TUNEL assays as well as DAPI and BrdU cell counts, recombinant human TFF3 significantly lowers retinoblastoma cell viability and increases apoptosis levels. Transient TFF3 overexpression likewise significantly increases RB cell apoptosis. Stable, lentiviral TFF3 overexpression lowers retinoblastoma cell viability, proliferation and growth and significantly increases cell death in retinoblastoma cells. Blockage experiments using a broad-spectrum caspase inhibitor and capase-3 immunocytochemistry revealed the involvement of caspases in general and of caspase-3 in particular in TFF3 induced apoptosis in retinoblastoma cell lines. Soft agarose and in ovo chicken chorioallantoic membrane (CAM) assays revealed that TFF3 overexpression influences anchorage independent growth and significantly decreases the size of tumors forming from retinoblastoma cells. Our study demonstrates that forced TFF3 expression exerts a significant pro-apoptotic, anti-proliferative, and tumor suppressive effect in retinoblastoma cells, setting a starting point for new additive chemotherapeutic approaches in the treatment of retinoblastoma.


Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , Trefoil Factor-3/physiology , Apoptosis , Blotting, Western , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Humans , Real-Time Polymerase Chain Reaction , Retinal Neoplasms/physiopathology , Retinoblastoma/physiopathology , Trefoil Factor-3/metabolism
6.
PLoS One ; 10(7): e0131467, 2015.
Article En | MEDLINE | ID: mdl-26173116

Retinoids have been shown to serve promising therapeutic agents for human cancers, e.g. the treatment of neuroblastoma. Synthetic retinoids, specific for particular retinoic acid (RA) receptors, are tested as new therapy strategies. In the present study, application of recombinant retinoic acid (RA) lowers retinoblastoma (RB) cell viability and induces apoptosis in RB cell lines. Combined treatment of RA and bone morphogenetic protein 4 (BMP-4) increases the pro-apoptotic effect of RA in the RB cells lines WERI-Rb1, Y-79, RB355, RBL-30 and RBL-15, indicating an additive effect. We could show that in WERI-Rb1 cells RA/BMP-4 mediated cell death is at least partially caspase-dependent, whereby RA and BMP-4 additively increased (i) Apaf-1 mRNA levels, (ii) caspase-9 cleavage activity and (iii) the number of activated, cleaved caspase-3 positive cells. Compared to single application of RA and BMP-4, combined RA/BMP-4 treatment significantly augments mRNA levels of the retinoic acid receptors (RARs) RARα and RARß and the retinoic X receptor (RXR) RXRγ suggesting an interaction in the induction of these RA receptor subtypes in WERI-Rb1 cells. Agonist studies revealed that both, RARs and RXRs are involved in RA/BMP-4 mediated apoptosis in WERI-Rb1 retinoblastoma cells. Employing specific RAR subtype antagonists and a RXRß and RXRγ knockdown, we proved that RA/BMP-4 apoptosis signaling in WERI-Rb1 cells requires the RA receptor subtypes RARα, RARß, RXRß and RXRγ. Deciphering signaling mechanisms underlying apoptosis induction of RA and BMP-4 in WERI-Rb1 cells, our study provides useful starting-points for future retinoid-based therapy strategies in retinoblastoma.


Apoptosis/drug effects , Bone Morphogenetic Protein 4/pharmacology , Retinoblastoma/pathology , Signal Transduction/drug effects , Tretinoin/pharmacology , Adult , Apoptotic Protease-Activating Factor 1/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Humans , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Up-Regulation/drug effects
7.
Nat Genet ; 47(8): 872-7, 2015 Aug.
Article En | MEDLINE | ID: mdl-26121086

Neuroblastoma is a malignancy of the developing sympathetic nervous system that is often lethal when relapse occurs. We here used whole-exome sequencing, mRNA expression profiling, array CGH and DNA methylation analysis to characterize 16 paired samples at diagnosis and relapse from individuals with neuroblastoma. The mutational burden significantly increased in relapsing tumors, accompanied by altered mutational signatures and reduced subclonal heterogeneity. Global allele frequencies at relapse indicated clonal mutation selection during disease progression. Promoter methylation patterns were consistent over disease course and were patient specific. Recurrent alterations at relapse included mutations in the putative CHD5 neuroblastoma tumor suppressor, chromosome 9p losses, DOCK8 mutations, inactivating mutations in PTPN14 and a relapse-specific activity pattern for the PTPN14 target YAP. Recurrent new mutations in HRAS, KRAS and genes mediating cell-cell interaction in 13 of 16 relapse tumors indicate disturbances in signaling pathways mediating mesenchymal transition. Our data shed light on genetic alteration frequency, identity and evolution in neuroblastoma.


Gene Expression Regulation, Neoplastic , Mutation , Neoplasm Recurrence, Local/genetics , Neuroblastoma/genetics , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Comparative Genomic Hybridization , DNA Copy Number Variations , DNA Helicases/genetics , Exome/genetics , Gene Expression Profiling/methods , Gene Frequency , Guanine Nucleotide Exchange Factors/genetics , Hippo Signaling Pathway , Humans , In Situ Hybridization, Fluorescence , Nerve Tissue Proteins/genetics , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Sequence Analysis, DNA/methods , Signal Transduction/genetics , Transcription Factors , YAP-Signaling Proteins
8.
Oncotarget ; 6(17): 15425-35, 2015 Jun 20.
Article En | MEDLINE | ID: mdl-26029996

Dysregulation of the cell cycle and cyclin-dependent kinases (cdks) is a hallmark of cancer cells. Intervention with cdk function is currently evaluated as a therapeutic option in many cancer types including neuroblastoma (NB), a common solid tumor of childhood. Re-analyses of mRNA profiling data from primary NB revealed that high level mRNA expression of both cdk1 and its corresponding cyclin, CCNB1, were significantly associated with worse patient outcome independent of MYCN amplification, a strong indicator of adverse NB prognosis. Cdk1 as well as CCNB1 expression were readily detectable in all embryonal tumor cell lines investigated. Pharmacological inhibition or siRNA-mediated knockdown of cdk1/CCNB1 induced proliferation arrest independent of MYCN status in NB cells. Sensitivity to cdk1 inhibition was modulated by TP53, which was demonstrated using isogenic cells with wild-type TP53 expressing either dominant-negative p53 or a short hairpin RNA directed against TP53. Apoptosis induced by cdk1 inhibition was dependent on caspase activation and was concomitant with upregulation of transcriptional targets of TP53. Our results confirm an essential role for the cdk1/CCNB1 complex in tumor cell survival. As relapsing embryonal tumors often present with p53 pathway alterations, these findings have potential implications for therapy approaches targeting cdks.


Cyclin B1/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Neuroblastoma/pathology , Rhabdomyosarcoma/pathology , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Apoptosis/genetics , CDC2 Protein Kinase , Caspase 8/metabolism , Caspase 9/metabolism , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cyclin B1/genetics , Cyclin-Dependent Kinases/genetics , Enzyme Activation/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Infant , N-Myc Proto-Oncogene Protein , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Quinolines/pharmacology , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering , Thiazoles/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
9.
Int J Cancer ; 136(10): 2293-303, 2015 May 15.
Article En | MEDLINE | ID: mdl-25348795

Previous studies have evaluated the role of miRNAs in cancer initiation and progression. MiR-34a was found to be downregulated in several tumors, including medulloblastomas. Here we employed targeted transgenesis to analyze the function of miR-34a in vivo. We generated mice with a constitutive deletion of the miR-34a gene. These mice were devoid of mir-34a expression in all analyzed tissues, but were viable and fertile. A comprehensive standardized phenotypic analysis including more than 300 single parameters revealed no apparent phenotype. Analysis of miR-34a expression in human medulloblastomas and medulloblastoma cell lines revealed significantly lower levels than in normal human cerebellum. Re-expression of miR-34a in human medulloblastoma cells reduced cell viability and proliferation, induced apoptosis and downregulated the miR-34a target genes, MYCN and SIRT1. Activation of the Shh pathway by targeting SmoA1 transgene overexpression causes medulloblastoma in mice, which is dependent on the presence and upregulation of Mycn. Analysis of miR-34a in medulloblastomas derived from ND2:SmoA1(tg) mice revealed significant suppression of miR-34a compared to normal cerebellum. Tumor incidence was significantly increased and tumor formation was significantly accelerated in mice transgenic for SmoA1 and lacking miR-34a. Interestingly, Mycn and Sirt1 were strongly expressed in medulloblastomas derived from these mice. We here demonstrate that miR-34a is dispensable for normal development, but that its loss accelerates medulloblastomagenesis. Strategies aiming to re-express miR-34a in tumors could, therefore, represent an efficient therapeutic option.


Cerebellar Neoplasms/pathology , Cerebellum/metabolism , Medulloblastoma/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Mice, Transgenic , Phenotype , Signal Transduction
10.
Oncotarget ; 5(22): 11180-92, 2014 Nov 30.
Article En | MEDLINE | ID: mdl-25361003

In neuroblastoma, the most common solid tumor of childhood, excellent prognosis is associated with extensive Schwann cell (SC) content and high-level expression of the neurotrophin receptor, NTRK1/TrkA, which is known to mediate neuroblastoma cell differentiation. We hypothesized that both stromal composition and neuroblastic differentiation are based on bidirectional neuroblastoma-SC interaction. Reanalysis of microarray data from human SY5Y neuroblastoma cells stably transfected with either NTRK1 or NTRK2 revealed upregulation of the mRNA for the SC growth factor, NRG1, in NTRK1-positive cells. Media conditioned by NTRK1-expressing neuroblastoma cells induced SC proliferation and migration, while antibody-based NRG1 neutralization significantly decreased these effects. Vice versa, NRG1-stimulated SC secreted the NTRK1-specific ligand, NGF. SC-conditioned medium activated the NTRK1 receptor in a neuroblastoma cell culture model conditionally expressing NTRK1 and induced differentiation markers in NTRK1-expressing cells. NTRK1 induction in neuroblastoma xenografts mixed with primary SC also significantly reduced tumor growth in vivo. We propose a model for NTRK1-mediated and NRG1-dependent attraction of adjacent SC, which in turn induce neuroblastic differentiation by secretion of the NTRK1-specific ligand, NGF. These findings have implications for understanding the mature and less malignant neuroblastoma phenotype associated with NTRK1 expression, and could assist the development of new therapeutic strategies for neuroblastoma differentiation.


Cell Communication/physiology , Membrane Glycoproteins/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Protein-Tyrosine Kinases/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Heterografts , Humans , Mice , Mice, Nude , Rats , Rats, Sprague-Dawley , Receptor, trkB , Transfection , Tumor Cells, Cultured , Up-Regulation
11.
Genes Chromosomes Cancer ; 50(5): 327-37, 2011 May.
Article En | MEDLINE | ID: mdl-21305643

In addition to mutations in both alleles of the retinoblastoma gene (RB1) alleles, retinoblastomas frequently show additional alterations including loss of chromosome arm 16q. In a previous study, the presence of 16q alterations was found to be associated with diffuse vitreous seeding of this tumor. This growth pattern is clinically important as it determines therapeutic decisions. The present study was designed to test this association and to narrow down the list of candidate genes in the minimal region of genomic loss on chromosome arm 16q. Our data confirm the association of 16q loss and diffuse vitreous seeding and define a minimal region of genomic loss of 6.6 Mb on 16q containing 86 known genes. As retinoblastoma is an embryonic tumor, we assumed that any gene relevant for its progression is likely to show regulated expression during retinogenesis. Microarray expression analysis of RNA from a continuous developmental series of murine retinas identified murine orthologs with regulated expression and these data helped to narrow the number of candidate genes in minimal region to 35. Analysis of gene expression in retinoblastomas with and without the loss of heterozygosity (LOH) on chromosome 16q further reduced this number to 26 candidate genes. One of these genes is cadherin 13 (CDH13) and notably, downregulation of CHD13 has previously been associated with poorer prognosis in various other cancers.


Chromosomes, Human, Pair 16 , Genes, Retinoblastoma , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Vitreous Body/pathology , Alleles , Animals , Cadherins/genetics , Chromosome Deletion , DNA, Neoplasm/blood , Gene Expression Regulation, Neoplastic , Genes, Regulator , Humans , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mutation , Polymorphism, Single Nucleotide , Retinal Neoplasms/blood , Retinal Neoplasms/pathology , Retinoblastoma/blood , Retinoblastoma/pathology , Sequence Deletion
12.
Int J Biol Sci ; 6(7): 700-15, 2010 Nov 24.
Article En | MEDLINE | ID: mdl-21152263

Bone morphogenetic proteins (BMPs) - expressed in the developing retina - are known to be involved in the regulation of cell proliferation and apoptosis in several tumor entities. The objective of this study was to determine the role of the BMP4 pathway in retinoblastoma cells, which are absent in a functional retinoblastoma (RB1) gene. BMP receptors were detected in all retinoblastoma cell lines investigated. A correct transmission of BMP signaling via the Smad1/5/8 pathway could be demonstrated in WERI-Rb1 retinoblastoma cells and application of recombinant human BMP4 resulted in an increase in apoptosis, which to a large extend is caspase independent. Cell proliferation was not affected by BMP4 signaling, although the pRb-related proteins p107 and p130, contributing to the regulation of the same genes, are still expressed. WERI-Rb1 cells exhibit elevated endogenous levels of p21(CIP1) and p53, but we did not detect any increase in p53, p21(CIP1)or p27(KIP1) expression levels. Id proteins became, however, strongly up-regulated upon exogenous BMP4 treatment. Thus, RB1 loss in WERI-Rb1 cells is obviously not compensated for by pRb-independent (e.g. p53-dependent) cell cycle control mechanisms, preventing an anti-proliferative response to BMP4, which normally induces cell cycle arrest.


Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , Retinoblastoma/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Immunohistochemistry , In Vitro Techniques , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 2/genetics , Retinoblastoma/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Smad7 Protein/genetics , Tumor Suppressor Protein p53/genetics
13.
Invest Ophthalmol Vis Sci ; 49(7): 3158-63, 2008 Jul.
Article En | MEDLINE | ID: mdl-18579764

PURPOSE: In contrast to the excellent survival rates of the malignant childhood tumor retinoblastoma (RB), morbidity is high in patients with this disease because of the enucleation or loss of retinal areas caused by current bulb-saving therapies. The authors aimed to preclinically assess the effects of photochemotherapy using second-generation photochemotherapeutics as a prerequisite to develop a promising therapeutic alternative. This therapy implies intravenous application of a photosensitizer activated locally by light of the appropriate wavelength. Activation leads to the formation of free radicals, vascular occlusion, and death of affected cells in the area of irradiation. The photosensitizer verteporfin is approved for the therapy of neovascularizations, such as age-related maculopathy. METHODS: The uptake of verteporfin in RB cell lines was investigated. Established RB cell lines, an RB subline resistant to etoposide, and dissociated cells from a primary RB were incubated with verteporfin and irradiated with activating laser light. Proliferation was measured at different time points after application. RESULTS: All five RB cell lines investigated incorporated verteporfin, and nanomolar concentrations were sufficient for effective killing. At lower doses, surviving cells started to proliferate again after several days, but verteporfin 50 ng/mL and 100 J/cm(2) were sufficient for irreversible killing. High verteporfin concentrations caused cell death with little to no irradiation. Etoposide-resistant cells and primary tumor cells had a comparable susceptibility to photodynamic therapy (PDT) as established parental cell lines. CONCLUSIONS: PDT using verteporfin efficiently kills chemotherapy-resistant and nonresistant retinoblastoma cell lines and primary tumor cells in vitro, and it warrants further preclinical evaluation as a therapeutic option for the treatment of retinoblastoma.


Photochemotherapy , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Cell Death , Cell Line, Tumor , Cell Proliferation , Cell Survival , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Etoposide/pharmacology , Humans , Osmolar Concentration , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/metabolism , Porphyrins/administration & dosage , Porphyrins/metabolism , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Retinal Neoplasms/physiopathology , Retinoblastoma/metabolism , Retinoblastoma/pathology , Retinoblastoma/physiopathology , Verteporfin
14.
Pediatr Blood Cancer ; 50(2): 218-22, 2008 Feb.
Article En | MEDLINE | ID: mdl-17973327

BACKGROUND: Neurotrophin receptor signaling regulates proliferation, differentiation and death of neuronal cells. Expression of Trk receptors has been implicated in the pathogenesis and prognosis of embryonal tumors, including neuroblastoma, nephroblastoma, and medulloblastoma. PROCEDURE: We analyzed TrkA, TrkB, TrkC, and p75 expression using semi-quantitative RT-PCR in 23 retinoblastomas and 8 retinoblastoma cell lines. Comparison of mRNA expression with clinical variables as well as the proliferation (PI) and apoptotic index (AI) of the tumor, was performed by Pearson correlation analysis and two-sample t-test. RESULTS: Almost all tumor samples and cell lines demonstrated high expression of all Trk receptors. Expression of TrkB and its ligand, BDNF, was most pronounced, suggesting TrkB to be the major Trk receptor involved in retinoblastoma biology. In contrast, p75 expression was substantially reduced in a subset of tumors and cell lines, in particular compared to its expression in normal retina. Tumors with infiltrative growth demonstrated significantly lower relative levels of TrkC expression than localized tumors (P = 0.004). High expression of TrkA was associated with a higher AI (P = 0.04), and high expression of TrkC was associated with a younger age of the patients (P = 0.03). Inhibition of Trk signaling by K252a resulted in marked growth inhibition of retinoblastoma cells in vitro. CONCLUSIONS: Our findings suggest a role for neurotrophin signaling in the biology of retinoblastoma. General Trk inhibitors are effective in decreasing growth rates of retinoblastoma cells in vitro, and should be evaluated in in vivo studies.


Receptors, Nerve Growth Factor/biosynthesis , Retinoblastoma/metabolism , Age Factors , Apoptosis/physiology , Brain-Derived Neurotrophic Factor/pharmacology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Humans , Immunohistochemistry , Infant , Nerve Growth Factor/pharmacology , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Nerve Growth Factor/biosynthesis , Receptor, Nerve Growth Factor/genetics , Receptor, trkA/biosynthesis , Receptor, trkA/genetics , Receptor, trkB/biosynthesis , Receptor, trkB/genetics , Receptor, trkC/biosynthesis , Receptor, trkC/genetics , Receptors, Nerve Growth Factor/genetics , Retinoblastoma/genetics , Retinoblastoma/pathology , Reverse Transcriptase Polymerase Chain Reaction
15.
Oncogene ; 24(42): 6441-9, 2005 Sep 22.
Article En | MEDLINE | ID: mdl-16007192

The paediatric eye tumour retinoblastoma is initiated by inactivation of RB1, a tumour suppressor on chromosome 13q. In addition to RB1 loss, many retinoblastomas show other genetic alterations including gains on chromosomes 6p21-pter and 1q31-q32. Recently, the minimal region of gains on chromosome 6 was narrowed to band p22. We examined genomic gains and expression changes in primary retinoblastomas to identify potential target genes in 6p22. Quantitative multiplex PCR detected copy numbers > or = 3 in 25 (33%) tumours and no gains in 31 of 76 (40%) tumours. The remaining 20 (26%) samples showed gains only at some loci, most often including E2F3 and DEK in 6p22.3. Analysis of RNA from 21 primary retinoblastomas showed that expression levels of these and some other genes in 6p22 correspond to DNA gains. However, KIF 13A, a reported candidate oncogene on 6p, was expressed at low levels or absent. Clinical manifestation of tumours with gains at all 6p22 loci was distinct in that distribution of age at diagnosis was markedly shifted to older age compared to tumours with no or partial gains. In summary, our results suggest that DEK and E2F3 are potential targets of 6p gains in retinoblastoma.


Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Human, Pair 6 , Oncogene Proteins/genetics , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Transcription Factors/genetics , Base Sequence , Blotting, Western , DNA Primers , E2F3 Transcription Factor , Gene Expression , Humans , Poly-ADP-Ribose Binding Proteins , Polymerase Chain Reaction
16.
Int J Cancer ; 116(4): 555-63, 2005 Sep 10.
Article En | MEDLINE | ID: mdl-15825178

Many retinoblastomas (Rbs) show genomic alterations in addition to mutational loss of both normal RB1 alleles. The most frequent of these changes are gains on chromosomes 1q and 6p and losses on 16q. To identify the genes targeted by gains on chromosome 1q, we used quantitative-multiplex PCR to determine DNA copy number changes in 76 primary tumors and 6 Rb cell lines. In addition, in 21 of these tumors, gene expression was analyzed by cDNA microarray hybridization. Increased copy numbers of loci on chromosome 1q were present in 34 (45%) primary tumors and in all 6 cell lines. Two regions of gain emerged, one in 1q32 and another in 1q21. Tumors with 1q gains showed higher RNA expression of several genes in these 2 regions. The clinical manifestation of tumors with and without gains was similar with regard to many aspects, including size, necrosis and calcification. However, the distribution of age at diagnosis was remarkably distinct, with earlier diagnosis in tumors without gains. This suggests that these tumors either are initiated earlier or grow faster than tumors with gains. This association with clinical manifestation indicates that gains on 1q are significant for the biology of Rb. The genes on 1q with copy number gains and overexpression are candidates that need to be tested for their individual contribution to the progression of Rb.


Chromosomes, Human, Pair 1 , Gene Dosage , Gene Expression Profiling , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Adolescent , Adult , Age of Onset , Aged , Child , Child, Preschool , Disease Progression , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA/biosynthesis
17.
J Mol Biol ; 315(4): 613-25, 2002 Jan 25.
Article En | MEDLINE | ID: mdl-11812134

The beta-thymosins are intracellular monomeric (G-)actin sequestering proteins forming 1:1 complexes with G-actin. Here, we analysed the interaction of thymosin beta(4) with F-actin. Thymosin beta(4) at 200 microM was chemically cross-linked to F-actin. In the presence of phalloidin, the chemically cross-linked actin:thymosin beta(4) complex was incorporated into F-actin. These mixed filaments were of normal appearance when inspected by conventional transmission electron microscopy after negative staining. We purified the chemically cross-linked actin:thymosin beta(4) complex, which polymerised only when phalloidin and the gelsolin:2-actin complex were present simultaneously. Using scanning transmission electron microscopy, the mass-per-length of control and actin:thymosin beta(4) filaments was found to be 16.0(+/-0.8) kDa/nm and 18.0(+/-0.9) kDa/nm, respectively, indicating an increase in subunit mass of 5.4 kDa. Analysis of the helical parameters revealed an increase of the crossover spacing of the two right-handed long-pitch helical strands from 36.0 to 40.5 nm. Difference map analysis of 3-D helical reconstruction of control and actin:thymosin beta(4) filaments yielded an elongated extra mass. Qualitatively, the overall size and shape of the difference mass were compatible with published data of the atomic structure of thymosin beta(4). The deduced binding sites of thymosin beta(4) to actin were in agreement with those identified previously. However, parts of the difference map might represent subtle conformational changes of both proteins occurring upon complex formation.


Actins/metabolism , Actins/ultrastructure , Thymosin/chemistry , Thymosin/metabolism , Actins/chemistry , Animals , Biopolymers/chemistry , Biopolymers/metabolism , Cross-Linking Reagents/metabolism , Gelsolin/metabolism , Humans , Kinetics , Microscopy, Electron , Microscopy, Electron, Scanning Transmission , Models, Molecular , Molecular Weight , Muscle, Skeletal/chemistry , Phalloidine/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Subunits , Rabbits
...