Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Cancer Lett ; 197(1-2): 151-5, 2003 Jul 18.
Article in English | MEDLINE | ID: mdl-12880975

ABSTRACT

Neuroblastoma (NB) is the most common extracranial solid tumor of childhood. In advanced disease stages, prognosis is poor and treatments have limited efficacy, thus novel strategies are warranted. The synthetic retinoid fenretinide (HPR) induces apoptosis in NB and melanoma cell lines. We reported an in vitro potentiation of HPR effects on melanoma cells when the drug is incorporated into GD2-targeted immunoliposomes (anti-GD2-SIL-HPR). Here, we investigated the antitumor activity of anti-GD2-SIL-HPR against NB cells, both in vitro and in vivo. Anti-GD2-immunoliposomes (anti-GD2-SIL) showed specific, competitive binding to, and uptake by, various NB cell lines. Moreover, anti-GD2-SIL-HPR presented increased selectivity and efficacy in inhibiting NB cell proliferation through the induction of apoptosis, compared to free drug and SL-HPR. In an in vivo NB metastatic model, we demonstrated that anti-GD2-SIL-HPR completely inhibited the development of macroscopic and microscopic metastases in comparison to controls. However, similar, but significantly less potent antitumor effect was observed also in mice treated with anti-GD2 immunoliposomes without HPR (anti-GD2-SIL-blank) or anti-GD2 mAb alone (P=0.0297 and P=0.0294, respectively, vs. anti-GD2-SIL-HPR). Moreover, our results clearly demonstrated that, although anti-GD2 mAb had a strong antitumor effect in this in vivo NB model, 100% curability was obtained only following treatment with anti-GD2-SIL-HPR (P<0.0001). Anti-GD2 liposomal HPR should receive clinical evaluation as adjuvant therapy of neuroblastoma.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Fenretinide/administration & dosage , Gangliosides/immunology , Neuroblastoma/drug therapy , Animals , Cell Division , Disease Models, Animal , Humans , Liposomes , Mice , Neuroblastoma/pathology , Tumor Cells, Cultured
2.
Oncogene ; 17(18): 2367-76, 1998 Nov 05.
Article in English | MEDLINE | ID: mdl-9811468

ABSTRACT

Functional nerve growth factor (NGF) responsiveness was investigated in human neuroblastoma (NB) cell lines in vitro and retinoic acid (RA) was found to transcriptionally enhance expression of the trkA, but not the trkB gene in GOTO cells, while the reverse was found in HTLA230 NB cells. Ciliary neurotrophic factor (CNTF) specifically induced trkA gene transcription in both cell lines. Transcriptional activation of the trkA gene increased total trkA protein and surface bound receptor, which was tyrosine phosphorylated upon NGF stimulation inducing immediate early response gene transcription (i.e. c-fos, Egr-1). Newly synthesized trkA protein had a molecular weight of 110 kDa and was post-translationally modified. Rapid down regulation of the receptor protein occurred upon NGF stimulation, despite the presence of high levels of trkA mRNA, due to an increased rate of receptor degradation. Transient DNA synthesis and cell proliferation upon NGF treatment occurred in GOTO cells with elevated trkA expression. In contrast, NGF induced neuronal differentiation in HTLA230 cells expressing the endogenous trkA receptor gene, despite the lack of p75 expression. Hence, transcriptional activation of trkA gene expression can be achieved by different signal pathways in human NB cells, but NGF can act either as mitogen or inducer of cell differentiation, depending on the tumor from which cells are derived.


Subject(s)
Immediate-Early Proteins , Nerve Growth Factors/pharmacology , Neuroblastoma/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Nerve Growth Factor/metabolism , Tretinoin/pharmacology , Ciliary Neurotrophic Factor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1 , Gene Expression Regulation, Neoplastic , Humans , Nerve Tissue Proteins , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Ciliary Neurotrophic Factor , Receptor, Nerve Growth Factor , Receptor, trkA , Receptors, Nerve Growth Factor/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
3.
Vis Neurosci ; 15(1): 87-95, 1998.
Article in English | MEDLINE | ID: mdl-9456508

ABSTRACT

The present study has identified a population of cone photoreceptors in the murine retina that are uniquely immunoreactive for protein kinase C (PKC). Wavelength-sensitive cone subtypes are segregated along the dorso-ventral axis in the mouse retina with ventral retina occupied exclusively by ultraviolet wavelength-sensitive (UVWS) cones, and dorsal retina dominated by middle wavelength-sensitive cones. PKC-positive cones are found primarily in the ventral retina, and double-label immunocytochemistry using a short wavelength-sensitive opsin antibody confirms that they specifically correspond to the UVWS cone subtype. The PKC antibody, as documented in other mammals, also identifies rod bipolar cells in the mouse retina. UVWS cones and bipolar cells have previously been shown to share transcriptional regulatory elements, as observed in transgenic mice encoding a portion of the human SWS-opsin promoter controlling the lacZ reporter gene. In such mice, the transgene product, beta-galactosidase, is expressed in populations of both cones and bipolar cells. The present study confirms that lacZ-expressing photoreceptors are indeed PKC-positive photoreceptors, but that the lacZ-expressing bipolar cells are not the PKC-positive rod bipolar cells. These cells must correspond to a type of cone bipolar cell.


Subject(s)
Interneurons/enzymology , Lipoproteins , Nerve Tissue Proteins , Protein Kinase C/metabolism , Retinal Cone Photoreceptor Cells/enzymology , Ultraviolet Rays , Animals , Antibodies, Monoclonal , Calbindins , Calcium-Binding Proteins/metabolism , Eye Proteins/metabolism , Fluorescent Antibody Technique, Indirect , Hippocalcin , Lac Operon , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recoverin , Retinal Cone Photoreceptor Cells/radiation effects , Rod Opsins/metabolism , S100 Calcium Binding Protein G/metabolism , beta-Galactosidase/metabolism
5.
Int J Cancer ; 67(3): 379-85, 1996 Jul 29.
Article in English | MEDLINE | ID: mdl-8707412

ABSTRACT

Neuroblastoma (NB), a neural crest derived tumor in children, shows a characteristic pattern of dissemination that includes adrenal glands, local lymph nodes, bone, liver, skin, and bone marrow. We have reconstructed a similar metastatic pattern in SCID mice following tail vein injection of human NB cells. HTLA230, an NB cell line isolated from a patient with advanced disease, and its NGF receptor (trkA) expressing derivative (18-10) cells, consistently disseminated to the liver, the adrenal gland, and the bone marrow, but not the lungs. Metastases in the different organs showed a characteristic hemorrhagic histopathology, and tumors in the bone marrow presented as syncytia-like cell aggregates, typically seen in patients. Cell lines reestablished from 18-10 derived liver and bone marrow metastases maintained their cellular morphology, growth behavior, N-myc overexpression, trkA expression, and functionally responded to NGF treatment, leading to growth arrest and neurite outgrowth. Hence circulating human NB cells in SCID mice show a similar organ-specific metastatic potential as seen in patients, independent of trkA expression.


Subject(s)
Neuroblastoma/pathology , Neuroblastoma/secondary , Adrenal Gland Neoplasms/secondary , Animals , Disease Models, Animal , Female , Humans , Liver Neoplasms, Experimental/secondary , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Neuroblastoma/ultrastructure , Organ Specificity , Ovarian Neoplasms/secondary , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/genetics , Transfection
6.
Oncogene ; 12(1): 77-86, 1996 Jan 04.
Article in English | MEDLINE | ID: mdl-8552402

ABSTRACT

Binding of nerve growth factor (NGF) to the TrkA receptor results in homodimerization, and activation of the intrinsic tyrosine kinase activity of the receptor, leading to multiple phosphorylations. We investigated the in vivo formation and composition of the receptor complex induced by NGF in a central nervous system-derived cell line (B104-neo), transfected with the human cDNA for TrkA. Using receptor-activated immunoprecipitation followed by analysis of the immune complexes by SDS-PAGE and immunoblotting, we show that NGF induces the association of TrkA with c-Crk-II in a multimeric complex that also includes SHC and PLC-gamma 1. While the tyrosine phosphorylation of TrkA, SHC and PLC-gamma 1 increased with time in the presence of NGF and was inhibited by the tyrosine kinase inhibitor K252a, the state of tyrosine phosphorylation of c-Crk-II did not appear to change with NGF treatment. Immunodepletion studies demonstrated that the interaction of c-Crk-II with TrkA not only occurs indirectly through the SHC proteins, but may also involve another mode of binding. Furthermore, we show that c-Crk-II is associated with tyrosine phosphorylated p130CAS in unstimulated cells and that NGF treatment results in the de-association of p130CAS/c-Crk-II complex in the absence of an apparent change in the tyrosine phosphorylation of p130CAS. These results clearly implicate c-Crk-II in the NGF signaling pathway and support the concept that more than one signaling molecule known to participate in the activation of Ras associates with TrkA upon NGF treatment.


Subject(s)
Nerve Growth Factors/pharmacology , Neurons/drug effects , Phosphoproteins/metabolism , Proteins , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Nerve Growth Factor/metabolism , Type C Phospholipases/metabolism , Cells, Cultured , Crk-Associated Substrate Protein , Humans , Neurons/metabolism , Phosphorylation , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-crk , Receptor Protein-Tyrosine Kinases/chemistry , Receptor, trkA , Receptors, Nerve Growth Factor/chemistry , Retinoblastoma-Like Protein p130 , Signal Transduction , src Homology Domains
7.
Oncogene ; 10(10): 1915-25, 1995 May 18.
Article in English | MEDLINE | ID: mdl-7761093

ABSTRACT

The effect of constitutively expressed N-myc gene on nerve growth factor (NGF) induced neuronal differentiation was investigated. B104, a rat central nervous system-derived cell line and its N-myc gene expressing derivative lines (C6, C7) (Bernards et al., 1986), were stably transfected with the trkA proto-oncogene and independent clones for each cell line were analysed. NGF induced phosphorylation of the trkA receptor, activated a cascade of cellular intermediaries such as phospholipase C gamma 1 and ERK proteins, and stimulated c-fos gene transcription in all trkA-expressing clones. NGF-mediated neuronal differentiation was observed solely in trkA-expressing B104-derived clones and was characterized by reduced cell growth, activation of NGF-regulated genes, and downregulation of the endogenous low-affinity NGF receptor gene (gp75NGFR). No such phenotypical changes occurred in trkA-expressing C6 or C7-derived clones following NGF treatment. These results are consistent with the hypothesis that constitutive expression of N-myc inhibits exit from cell cycle and blocks neuronal cell differentiation.


Subject(s)
Genes, myc/physiology , Nerve Growth Factors/pharmacology , Neurons/cytology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line , Gene Expression Regulation/drug effects , Genes, fos/physiology , Membrane Glycoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rats , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Nerve Growth Factor , Receptor, trkA , Receptors, Nerve Growth Factor/genetics
8.
Proc Natl Acad Sci U S A ; 91(7): 2611-5, 1994 Mar 29.
Article in English | MEDLINE | ID: mdl-8146162

ABSTRACT

Transgenic mouse lines were generated using either 3.8 or 1.1 kb of 5' upstream flanking sequence from the human blue opsin gene fused to the lacZ or human growth hormone reporter gene. Mice were analyzed for appropriate cell-specific and developmental expression patterns. In 13 independently derived lines of animals, transgene expression was limited to photoreceptor and inner nuclear layer cells. Photoreceptors were identified as cone cells based on morphological criteria and colocalization of transgene expression with the cone-associated marker, peanut agglutinin lectin. More specifically, transgene-positive photoreceptors were identified as short-wave cone cells (S-cones) by using the short-wave color opsin-specific antibody, OS-2. Reporter-gene-positive cells of the inner nuclear layer were identified as bipolar cells based on morphological criteria. Transgenes and the endogenous mouse short-wave opsin gene were transcriptionally coactivated at embryonic day 13. These results show that 3.8 or 1.1 kb of human blue opsin upstream flanking sequences are capable of directing expression in short-wave cone cells in a spatially and temporally appropriate fashion and that the human blue opsin gene is the homologue of the short-wave-sensitive pigment, S-opsin, in the short-wave cones of the mouse retina. Expression in the bipolar cells may reflect regulatory mechanisms that are common to these cells and to the cone photoreceptors.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic/genetics , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Rod Opsins/genetics , Animals , Base Sequence , Genes, Reporter , Growth Hormone/biosynthesis , Growth Hormone/genetics , Histocytochemistry , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Molecular Sequence Data , Retina/cytology , Retina/embryology , Rod Opsins/biosynthesis , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
10.
Mol Cell Biol ; 13(12): 7447-56, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8246962

ABSTRACT

The human trkA cDNA was transfected into a malignant human neuroblastoma (NB) cell line (HTLA230) to investigate its role in NB growth and differentiation. This cell line lacks expression of both endogenous trkA and gp75NGFR genes. Transfectants expressing the trkA mRNA and surface-bound receptors transcriptionally activate immediate-early genes (c-fos, c-jun, and jun-B) following nerve growth factor (NGF) stimulation. NGF treatment induces growth arrest as well as down-regulation of the amplified N-myc oncogene. Genes selectively expressed in mature neurons (SCG-10, ret proto-oncogene, GAP-43, etc.) are transcriptionally activated, and neurite outgrowth further demonstrates differentiation of transfectants following NGF stimulation. trkA-expressing NB cells remain tumorigenic in nude mice; however, subcutaneous treatment of tumor-bearing mice with NGF induces Schwannian and neuronal cell differentiation similar to the induction seen in human ganglioneuroblastomas. Thus, trkA expression in HTLA230 cells is sufficient to generate a functional NGF receptor complex that leads to growth-arrested and differentiated NB cells in vitro and in vivo in the presence of NGF. Hence, NGF may play a crucial role in NB cell differentiation and regression in vivo.


Subject(s)
DNA, Complementary/genetics , DNA, Neoplasm/genetics , Neuroblastoma/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Nerve Growth Factor/genetics , Animals , Cell Differentiation/genetics , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Nerve Growth Factors/pharmacology , Neuroblastoma/pathology , Proto-Oncogene Mas , Receptor, trkA , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
11.
J Natl Cancer Inst ; 85(5): 377-84, 1993 Mar 03.
Article in English | MEDLINE | ID: mdl-8433391

ABSTRACT

BACKGROUND: Neuroblastoma is a malignancy of the sympathetic nervous system. Nerve growth factor, which has a major role in development of the sympathetic nervous system, has high-affinity (gp140TRK-A) and low-affinity (gp75NGFR) cell-surface receptors. We recently reported preliminary study results showing a lack of gp140TRK-A receptors and rapid disease progression in neuroblastomas, particularly those with amplification of the N-myc (also known as MYCN) proto-oncogene. PURPOSE: This retrospective study was designed to determine if expression of nerve growth factor receptor messenger RNA (mRNA) was associated with biologic and clinical parameters and with survival in neuroblastoma. METHODS: We obtained 80 untreated primary neuroblastomas that had been snap-frozen and stored after surgical excision. To determine expression of gp140TRK-A and gp75NGFR, we performed Northern blot analyses on total RNA from the specimens. Samples from the same specimens were examined for N-myc proto-oncogene amplification, RNA expression, and histologic differentiation, and clinical stage at diagnosis and survival were determined. RESULTS: Of the 80 neuroblastomas, 65 (81%) expressed gp140TRK-A RNA. However, three (27%) of the 11 tumors with genomic amplification and high expression of N-myc RNA and 62 (90%) of the 69 without genomic amplification or detectable N-myc RNA expressed gp140TRK-A mRNA. The inverse relationship between gp140TRK-A mRNA and N-myc expression had high statistical significance (P < .0001). Of the 67 tumors assessable for histologic differentiation, the 13 lacking gp140TRK-A mRNA were histologically undifferentiated, whereas 19 (35%) of the 54 expressing it were differentiated (P = .041). Only 10 (53%) of the 19 metastatic (stage IV) tumors expressed gp140TRK-A mRNA, compared with 90% for other stages (P = .0003). Survival 2 years after diagnosis was 92%, 78%, and 14% for patients whose tumors expressed high, intermediate, and no gp140TRK-A mRNA, respectively (P < .0001). Univariate and multivariate analyses demonstrated that N-myc and gp140TRK-A expression of mRNA and clinical staging were independent predictors of survival. Expression of gp75NGFR mRNA did not correlate with gp140TRK-A mRNA expression, histologic differentiation, stage, or survival. CONCLUSIONS: The expression of gp140TRK-A mRNA correlates with distinct biologic and clinical subsets of neuroblastoma, which suggests a role for the high-affinity nerve growth factor receptors in determining the phenotype of neuroblastoma. The absence of gp140TRK-A mRNA expression, whether or not the N-myc proto-oncogene is amplified, is associated with tumor progression.


Subject(s)
Neuroblastoma/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Receptors, Nerve Growth Factor/metabolism , Humans , Neoplasm Staging , Neuroblastoma/genetics , Neuroblastoma/mortality , Neuroblastoma/pathology , Prognosis , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Receptor, trkA , Receptors, Nerve Growth Factor/genetics , Retrospective Studies , Survival Analysis
12.
Int J Cancer ; 52(2): 298-304, 1992 Sep 09.
Article in English | MEDLINE | ID: mdl-1325952

ABSTRACT

The human sarcoma cell line HT1080 was found, by in situ hybridization, to consist of cells expressing various levels of urokinase (uPA) and tissue type (tPA) plasminogen activator (PA) suggesting clonal variation of expression of these genes. Colonies originating from single HT1080 cells were, therefore, established and screened for PA activity using a fibrin agarose overlay. Colonies inducing lysis (clone C+ and H+) or no lysis (clones B- and M-) were isolated and tested for mRNA levels of uPA, tPA, uPA receptor (uPAR) and the 3 PA inhibitors (PAI), PAI-1, PAI-2 and protease-nexin I. The different clones revealed considerable variation of expression of the different PA and PAI genes, with lysis-inducing clones expressing mainly the PA genes, whereas non-lysing clones demonstrated higher expression of the PAI genes. Amplification or loss of specific genes was excluded by Southern blotting. The protein levels of cellular and secreted PA and PAI determined by ELISA and Western blots demonstrated a pattern similar to that observed for PA and PAI mRNA concentrations, suggesting clonal differences either on the level of transcription or in RNA processing and/or stability. Due to complex interactions between PA and PAI, neither mRNA nor protein levels of the different genes were predictive for the amount of functional PA activity present in the supernatant or on the cell surface of the different clones. Receptor-bound uPA activity was found to be considerably higher in lysis-inducing than in non-lysing clones and the activity was dependent on neutralization by PAI-1 rather than on the level of uPAR mRNA.


Subject(s)
Gene Expression Regulation, Neoplastic , RNA, Messenger/analysis , Receptors, Cell Surface/genetics , Sarcoma/genetics , Tissue Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/genetics , Fibrinolysin/analysis , Gene Expression Regulation, Enzymologic , Humans , RNA, Neoplasm/analysis , Receptors, Urokinase Plasminogen Activator , Tissue Plasminogen Activator/antagonists & inhibitors , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
13.
Int J Cancer ; 51(5): 727-32, 1992 Jul 09.
Article in English | MEDLINE | ID: mdl-1612781

ABSTRACT

The effect of a complex in vitro synthesized extracellular matrix (ECM) and its components on growth and phenotypical differentiation of a human neuroblastoma (NB) cell line (HTLA230) was investigated. Rat smooth-muscle-cell (R22CIF)-derived ECM composed of collagen, glycoproteins, and glycosaminoglycans (GAGs) promoted spontaneous neurite outgrowth of HTLA230 cells but did not alter their growth kinetic or cloning efficiency as compared with cells seeded onto gelatin-coated dishes. The matrix significantly enhanced, quantitatively and qualitatively, the responsiveness of HTLA230 cells to retinoic acid (RA), and a substantially reduced growth rate was observed in the presence of RA with cells grown on the ECM. Biochemical modification of the composition of the R22CIF-matrix by trypsin digestion and/or high-salt extraction (4 M guanidinium) demonstrated that the ratio of chondroitin sulfate to hyaluronic acid (HA) present in the ECM determines the capacity of the matrix to promote NB differentiation. A human fibroblast (T-1)-derived ECM, which has a biochemical composition of the GAG component similar to that of the trypsinized R22CIF-matrix, but which has a high amount of glycoproteins, confirmed these results. Nerve-growth-factor (NGF)-induced differentiation in a variant HTLA 230 cell line was inhibited when cells were grown on an ECM with a low ratio of chondroitin sulfate/HA. The composition of the ECM thus modulates the responsiveness to various differentiation-inducing agents and alters the phenotype of NB cells.


Subject(s)
Extracellular Matrix/physiology , Neuroblastoma/pathology , Animals , Cell Differentiation , Clone Cells/pathology , Collagen/pharmacology , Glycoproteins/pharmacology , Glycosaminoglycans/pharmacology , Humans , Neurites/pathology , Phenotype , Rats , Tretinoin/pharmacology , Trypsin/metabolism , Tumor Cells, Cultured
14.
Int J Cancer ; 51(2): 250-8, 1992 May 08.
Article in English | MEDLINE | ID: mdl-1568793

ABSTRACT

We have isolated a human neuroblastoma (NB) cell line, HTLA230, from the bone-marrow aspirate of a patient with stage-IV disease. Subcutaneous tumors after inoculation of HTLA230 cells into nude mice were composed of primitive neuroblasts which rarely contained neuro-secretory granules. Cytogenetic studies of the cell line demonstrated 2 distinct populations of cells with common chromosomal markers. Stable sub-clones with a differentiated or undifferentiated cell morphology were isolated, demonstrating phenotypical heterogeneity of the HTLA230 parental cell line. Treatment with retinoic acid (RA) induced extensive neurite outgrowth in the parental cell line and in phenotypically differentiated sub-clones, but rarely in undifferentiated ones. Long-term treatment with RA was not associated with down-modulation of mycN-gene expression, which could be achieved only in cultures treated additionally with aphidicolin, a DNA-synthesis inhibitor, thus eliminating growing NB cells. A RA resistant subclone (CI-5) was isolated from parental HTLA230 cells grown at clonal cell density. Cells originally showed a homogeneously differentiated morphology; however, flat cells (F-cells) appeared with time and were subsequently separately propagated. Transdifferentiation of isolated F-cells into cells with neuron-like (N-cell) morphology was observed. Immunohistochemical analysis demonstrated that F-cells had lost the expression of neuronal markers, including HNK-I and A2B5, and expressed the intermediate filament, vimentin. Furthermore, F-cells showed high incorporation of [methyl-3H] thymidine (3H-TdR) by autoradiography but no mycN protein could be detected, although present in the parental cell line. These results then suggest that the isolated NB cell line and the RA-resistant variant line represent an excellent in vitro model with which the bi-modal differentiation pathway of NB can be analyzed on a molecular biological level.


Subject(s)
Cell Differentiation/physiology , Neuroblastoma/pathology , Animals , Cell Differentiation/drug effects , Drug Resistance , Gene Amplification , Gene Expression Regulation, Neoplastic/genetics , Genes, myc/genetics , Humans , Infant , Male , Mice , Mice, Nude , Neuroblastoma/genetics , Neuroblastoma/ultrastructure , Nucleic Acid Hybridization , Phenotype , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Tretinoin/pharmacology , Tumor Cells, Cultured
15.
Cancer Genet Cytogenet ; 56(1): 31-8, 1991 Oct 01.
Article in English | MEDLINE | ID: mdl-1747867

ABSTRACT

The malignant rhabdoid tumor is a rare, poorly understood tumor which occurs primarily in children. The kidney is a frequent primary site of origin, but the tumor has arisen in other mesodermally derived tissues as well. Controversy exists regarding the embryonic origin of the rhabdoid tumor and recent histopathologic studies suggest that it may be of neuroepithelial origin. Our immunohistochemical and electron micrographic studies support this theory. No consistent chromosome abnormalities have been reported in this tumor and no cell lines are available for study. We have established and characterized the first rhabdoid tumor cell line. It possesses a specific chromosomal abnormality, 46,XY,t(11;22)(p15.5;q11.23). The translocation may provide an important clue to the pathogenesis of the tumor as well as an opportunity for further study of the involved chromosome regions.


Subject(s)
Chromosome Aberrations , Chromosome Disorders , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 22 , Rhabdomyosarcoma/genetics , Soft Tissue Neoplasms/genetics , Translocation, Genetic , X Chromosome , Y Chromosome , Adult , Cell Line , Chromosome Banding , Humans , Karyotyping , Male , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/ultrastructure , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/ultrastructure
17.
Mol Cell Biol ; 10(9): 5015-20, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2167446

ABSTRACT

Human nerve growth factor (NGF) receptor (NGFR) cDNA was transfected into a neuroblastoma cell line (HTLA 230) which does not express a functional NGF-NGFR signal transduction cascade. Short-term treatment of stably transfected cells (98-3) expressing membrane-bound NGF receptor molecules resulted in a cell cycle-dependent, transient expression of the c-fos gene upon treatment with NGF, suggesting the presence of functional high-affinity NGFR. Extensive outgrowth of neurites and cessation of DNA synthesis occurred in transfectants grown on an extracellular matrix after long-term treatment with NGF, suggesting terminal differentiation. Our data support the idea that introduction of a constitutively expressed NGFR cDNA into cells with neuronal background results in the assembly of a functional NGF-NGFR signal cascade in a permissive extracellular environment.


Subject(s)
Nerve Growth Factors/pharmacology , Neurons/cytology , Receptors, Cell Surface/physiology , Tumor Cells, Cultured/cytology , Animals , Autoradiography , Cell Differentiation , Cell Line , DNA, Neoplasm/genetics , Flow Cytometry , Humans , Melanoma , Nerve Growth Factors/metabolism , Neuroblastoma , Neurons/drug effects , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Receptors, Cell Surface/genetics , Receptors, Nerve Growth Factor , Signal Transduction , Thymidine/metabolism , Transfection , Tritium , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
18.
J Clin Invest ; 85(6): 1872-8, 1990 Jun.
Article in English | MEDLINE | ID: mdl-2161431

ABSTRACT

Retinoblastoma is a malignant intraocular tumor that primarily affects small children. These tumors are primitive neuroectodermal malignancies, however some of them show morphologic evidence of differentiation into photoreceptors. Phototransduction cascades are a series of biochemical reactions that convert a photon of light into a neural impulse in rods and cones. The components of these cascades are uniquely expressed in photoreceptors and, although functionally similar, distinct components of these cascades are expressed in rods and cones. Using HPLC anion exchange chromatography, Western blot analysis, and specific monoclonal and polyclonal antibodies, we found that the cone but not the rod cGMP phosphodiesterase is functionally expressed in all six primary retinoblastomas examined and in three continuous retinoblastoma cell lines. Morphologic evidence of differentiation did not correlate with the expression of the enzyme. Furthermore, GTP analogues could activate the phosphodiesterase activity suggesting that an intact phototransduction cascade is present in the tumors. The presence of the cone phototransduction cascade in retinoblastoma confirms that this tumor has biochemically differentiated along the cone cell lineage.


Subject(s)
Photoreceptor Cells/physiology , Retinoblastoma/physiopathology , 3',5'-Cyclic-GMP Phosphodiesterases/physiology , Blotting, Western , Calmodulin/physiology , Cell Differentiation , Enzyme Activation , GTP-Binding Proteins/physiology , Humans , Molecular Weight , Photoreceptor Cells/cytology , Retina/enzymology , Retinoblastoma/pathology , Vision, Ocular
19.
Proc Natl Acad Sci U S A ; 87(7): 2775-9, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2181449

ABSTRACT

We have used polyclonal anti-synthetic peptide serum to study the role of retinoblastoma gene (RB) inactivation in a variety of human tumor cell lines. Our analysis indicates that inactivation of the RB protein, p105-Rb, is universal in retinoblastoma cells, vindicating the predictions of the Knudson "two-hit" hypothesis. In addition, our analysis has shown that inactivations of the RB gene are nearly as frequent in a more common human tumor, small cell lung carcinoma. One-third of bladder carcinomas surveyed also carry altered or absent p105-Rb. Other human tumors by contrast demonstrate only infrequent inactivation of the RB gene. These results suggest that inactivation of the RB gene is a critical step in the pathogenesis of a subset of human tumors.


Subject(s)
Eye Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Oncogenes , Phosphoproteins/genetics , Retinoblastoma/genetics , Base Sequence , Blotting, Southern , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Humans , Methionine/metabolism , Molecular Sequence Data , Mutation , Polymerase Chain Reaction , Protein Biosynthesis , RNA, Messenger/genetics , Retinoblastoma Protein , Transcription, Genetic , Tumor Cells, Cultured/metabolism
20.
Mol Cell Biol ; 9(11): 4722-30, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2601695

ABSTRACT

Several human rhabdomyosarcoma cell lines, cultured primary tumor explants, and biopsies of tumor and normal skeletal muscle tissue expressed a 2.0-kilobase transcript that hybridized to the mouse muscle determination gene MyoD1. This transcript was found in tumor cell lines and primary explants that developed multinucleated myotubes but was absent in Wilms' tumors or cell lines and primary explants that developed multinucleated myotubes but was absent in Wilms' tumors or cell lines derived from other mesenchymal tumor cell types. Expression of the human homolog of MyoD1 therefore can define a tumor as a rhabdomyosarcoma. Transfection of the mouse MyoD1 gene into the human rhabdomyosarcoma cell line RD increased the ability of the tumor cells to differentiate into multinucleated myotubes and enhanced myosin heavy-chain gene expression but did not decrease tumorigenicity in nude mice.


Subject(s)
Cell Transformation, Neoplastic , Gene Expression , Genes , Muscles/cytology , Rhabdomyosarcoma/genetics , Animals , Blotting, Northern , Blotting, Southern , Carcinogenicity Tests , Cell Differentiation , Cell Line , Humans , Mice , Myosins/analysis , Myosins/genetics , Rhabdomyosarcoma/pathology , Transcription, Genetic , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...