Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Curr Med Chem ; 19(36): 6214-23, 2012.
Article in English | MEDLINE | ID: mdl-22664252

ABSTRACT

MicroRNAs (miRNA) are a recently recognised class of small, non-coding RNAs involved in the post-transcriptional regulation of gene expression and with crucial implication for mammalian development. In particular, they play key roles in neuronal development, from early neurogenesis to neuronal differentiation and synaptic development, and also in in vitro systems. The detection of embryotoxic hazards in the preclinical phase is still a challenge, often due to species-species variations. In this study we analysed whether miRNA expression profiles in a human pluripotent cell model can be a helpful tool for a more mechanistic approach to pharmacology and toxicology. Differentiating human pluripotent cells were repeatedly treated with non-cytotoxic doses of methylmercury chloride (MeHgCl), a well known brain developmental toxicant. The expression of proteins, mRNA and miRNAs were used to monitor successful neural differentiation. Significant changes in the expression of 12 miRNAs were detected. By using available bioinformatics tools, we obtained validated and predicted targets for the identified miRNAs, on which we performed functional clustering analysis. Through this approach, we identified several terms and functional clusters associated with neural development, together with indicators of general toxic effect, such as apoptosis or stress response-related genes. Interestingly, our results also suggest a previously undiscovered association between MeHgCl and the ubiquitin-proteasome protein degradation pathway. Although further investigations are needed, our results suggest that miRNA expression analysis is a powerful tool in pathway-oriented toxicity and could improve early-phase hazard assessments.


Subject(s)
Gene Expression Profiling , MicroRNAs/metabolism , Models, Biological , Neurons/cytology , Cell Line , Cluster Analysis , Computational Biology , Humans , Methylmercury Compounds/chemistry , Methylmercury Compounds/toxicity , Neurogenesis/drug effects , Neurons/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
2.
Toxicology ; 290(1): 50-8, 2011 Nov 28.
Article in English | MEDLINE | ID: mdl-21871943

ABSTRACT

The application of toxicogenomics as a predictive tool for chemical risk assessment has been under evaluation by the toxicology community for more than a decade. However, it predominately remains a tool for investigative research rather than for regulatory risk assessment. In this study, we assessed whether the current generation of microarray technology in combination with an in vitro experimental design was capable of generating robust, reproducible data of sufficient quality to show promise as a tool for regulatory risk assessment. To this end, we designed a prospective collaborative study to determine the level of inter- and intra-laboratory reproducibility between three independent laboratories. All test centres (TCs) adopted the same protocols for all aspects of the toxicogenomic experiment including cell culture, chemical exposure, RNA extraction, microarray data generation and analysis. As a case study, the genotoxic carcinogen benzo[a]pyrene (B[a]P) and the human hepatoma cell line HepG2 were used to generate three comparable toxicogenomic data sets. High levels of technical reproducibility were demonstrated using a widely employed gene expression microarray platform. While differences at the global transcriptome level were observed between the TCs, a common subset of B[a]P responsive genes (n=400 gene probes) was identified at all TCs which included many genes previously reported in the literature as B[a]P responsive. These data show promise that the current generation of microarray technology, in combination with a standard in vitro experimental design, can produce robust data that can be generated reproducibly in independent laboratories. Future work will need to determine whether such reproducible in vitro model(s) can be predictive for a range of toxic chemicals with different mechanisms of action and thus be considered as part of future testing regimes for regulatory risk assessment.


Subject(s)
Databases, Genetic/standards , Laboratories/standards , Research Design/standards , Toxicogenetics/standards , Hep G2 Cells , Humans , Principal Component Analysis/methods , Principal Component Analysis/standards , Prospective Studies , Protein Array Analysis/methods , Protein Array Analysis/standards , Reproducibility of Results , Toxicogenetics/methods
3.
Exp Toxicol Pathol ; 61(1): 33-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18650076

ABSTRACT

Volatile organic compounds (VOCs) such as benzene and toluene, and low molecular weight carbonyls like formaldehyde belong to the main air pollutants found in indoor environments. They are suspected to induce acute and chronic adverse health effects like asthma, allergic and cardiovascular diseases, and strongly affect well-being. Our aim was to further develop and optimize an in vitro method to study the exposure of epithelial tumour lung cells (A549) by using a commercial exposure chamber (CULTEX) to assess the biological effects of VOCs and carbonyl compounds at low concentration levels. Exposing the cells to toluene, benzene and formaldehyde at mixing ratios varying from 0.1 to 0.6ppmv in air resulted in reproducible direct effects with the induction of an inflammatory response and a modification of the glutathione redox status.


Subject(s)
Cell Culture Techniques/methods , Respiratory Mucosa/drug effects , Volatile Organic Compounds/toxicity , Air Pollution, Indoor/adverse effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/pathology , Glutathione/metabolism , Humans , Inflammation/chemically induced , Interleukin-8/metabolism , L-Lactate Dehydrogenase/metabolism , Respiratory Mucosa/metabolism
4.
Exp Toxicol Pathol ; 61(4): 381-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19046626

ABSTRACT

Human epithelial lung cells (A549) were exposed to toluene and benzene in the air as individual compounds and mixtures at concentrations of about 0.25ppmv in a specifically adapted fumigation device. Possible early toxicological effects at cellular level have been determined by lactate dehydrogenase (LDH), glutathione redox status (GSH) and comet assay. An hour of exposure to 0.25ppmv of toluene in the air induced DNA damages which were repaired within 24h after the treatment. No DNA damage was detected by applying a similar concentration of benzene, but there was a decrease in the glutathione ratio. Exposure to a mixture of toluene and benzene in air led to an increase in the cytotoxic effect and DNA damage without any further repair, but did not induce any changes in the glutathione redox status.


Subject(s)
Air Pollution, Indoor/adverse effects , Benzene/toxicity , Epithelial Cells/drug effects , Lung/drug effects , Toluene/toxicity , Benzene/chemistry , Cell Line , Cell Survival/drug effects , Comet Assay , DNA Damage , DNA Repair , Drug Synergism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glutathione/metabolism , Humans , Lung/cytology , Oxidation-Reduction , Toluene/chemistry
5.
Cell Prolif ; 39(6): 611-22, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17109643

ABSTRACT

A new murine cell line, named GFPneu, was established from a mammary adenocarcinoma arising in double transgenic MMTVneu x CMV-GFP mice. Breast tumours develop in 100% of females after 2 months latency, as a result of the over-expression of the activated rat neu oncogene in the mammary glands. All tissues, and in particular the breast tumours, express the GFP protein. This cell line was tumorigenic when inoculated into nude mice and the derived tumours showed the same histological features as the primaries from which they were isolated. Their histopathology reproduces many characteristics of human breast adenocarcinomas, in particular their ability to metastasize. The GFP marker allows us to visualize the presence of lung metastases in fresh tissues immediately, to confirm the histopathology. From a lung metastatic fluorescent nodule, we derived a further cell line, named MTP-GFP, which we also characterized. These two cell lines could be useful to study the role played by the neu oncogene in the maintenance of the transformed phenotype, in the metastatic process, to test novel therapeutic strategies to inhibit primary tumour growth and to observe the generation of distant metastases.


Subject(s)
Adenocarcinoma/genetics , Cell Line, Tumor , Genes, erbB-2/genetics , Green Fluorescent Proteins/genetics , Mammary Neoplasms, Animal/genetics , Adenocarcinoma/secondary , Animals , Female , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/pathology , Mice , Mice, Nude , Mice, Transgenic , Telomere
6.
Gene Ther ; 10(22): 1903-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14502219

ABSTRACT

Treatments available to women with locally advanced breast cancer are unsatisfactory, since most patients succumb to metastatic spread. Therefore, there is a need to devise novel therapeutic combinations that effectively inhibit metastatization and to test them in animal models of breast cancer showing strong similarities with their human counterpart, including the ability to give rise to metastases. With these considerations in mind, tamoxifen (TAM), 4-hydrotamoxifen (4-HT) or liposome-complexed DNA constructs coding for antiangiogenic/anti-invasion proteins (angiostatin, TIMP-2, IFN-alpha(1), sFLT-1) were individually administered to MMTVneu transgenic mice. Significant inhibition of primary tumor growth was obtained with TAM (40% inhibition, P=0.049), angiostatin (85% inhibition, P=0.001) and TIMP-2 (60% inhibition, P=0.015). No lung metastasis was observed in any of these treated mice at 5 months, compared with a rate of 70% in control groups. These observations were the basis for designing a combined treatment with all these compounds. The association of angiostatin, TIMP-2 and TAM was greatly effective at the primary tumor level (90% inhibition, P=0.01). Moreover, all the mice treated with this association were metastasis free at a time point (6 months) in which seven out of nine control mice were either dead from disseminated cancer or showed lung metastasis. This combined therapy could become an important component of anticancer therapy in humans.


Subject(s)
Breast Neoplasms/therapy , Estrogen Receptor Modulators/therapeutic use , Genetic Therapy/methods , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Tamoxifen/therapeutic use , Angiogenesis Inhibitors/genetics , Angiostatins , Animals , Combined Modality Therapy , Extracellular Matrix Proteins/genetics , Female , Gene Expression , Genes, erbB-2 , Genetic Vectors/administration & dosage , Interferon-alpha/genetics , Mice , Mice, Transgenic , Models, Animal , Myosin Heavy Chains , Nonmuscle Myosin Type IIB , Peptide Fragments/genetics , Plasminogen/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics
7.
Gene Ther ; 9(19): 1338-41, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12224018

ABSTRACT

Breast tumor growth and metastasization are both hormone-sensitive and angiogenesis-dependent. Recent work carried out in our laboratory on a transgenic model of breast cancer displaying many similarities to its human counterpart, has shown that liposome-mediated angiostatin cDNA delivery partially inhibits both local and metastatic growth. However, it is now recognized that anti-angiogenesis strategy alone cannot completely arrest tumor growth and spread, and this led to the suggestion that approaches based on different molecular mechanisms could usefully be combined. In the present work, we investigated whether tamoxifen, a classical antiestrogen agent widely used in human therapy, could improve the results obtained with angiostatin alone. Further reduction of local growth was achieved with the combined regimen with respect to angiostatin or tamoxifen alone, while, as expected, no metastatic growth was detected in either group. We therefore conclude that a combination of angiogenesis inhibitors with antiestrogen drugs might be useful in humans and that other associations between conventional and gene transfer-mediated therapy are worth investigating and will soon become important components of anticancer therapy.


Subject(s)
Genetic Therapy/methods , Mammary Neoplasms, Experimental/therapy , Neovascularization, Pathologic/prevention & control , Peptide Fragments/genetics , Plasminogen/genetics , Tamoxifen/therapeutic use , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Angiostatins , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Combined Modality Therapy , DNA, Complementary/genetics , Female , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Transgenic , Peptide Fragments/metabolism , Plasminogen/metabolism
8.
Cancer Gene Ther ; 8(7): 491-6, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11498770

ABSTRACT

The effect of local and systemic delivery of the angiostatin gene on human melanoma growth was studied in nude mice. Liposome-coated plasmids carrying the cDNA coding for murine and human angiostatin (CMVang and BSHang) were injected weekly, locally or systemically, in mice transplanted with melanoma cells. The treatment reduced melanoma growth by 50% to 90% compared to that occurring in control animals treated with liposome-coated plasmid carrying the lacZ gene or in untreated controls. The growth of both locally injected and controlateral uninjected tumors in mice bearing two melanoma grafts was significantly suppressed after intratumoral treatment. Tumor growth inhibition was also observed in mice treated by intraperitoneal delivery, suggesting that angiostatin gene therapy acts through a systemic effect. Both melanoma growth suppression and delay in the onset of tumor growth were observed in treated mice. PCR performed on tumors and normal tissues showed that the lipofected DNA was present in tissues from treated mice, and angiostatin expression was demonstrated by RT-PCR. Histopathological analysis of melanoma nodules revealed an increase in apoptotic cells and a reduction in vessel density in tumors from treated mice. Our results suggest that systemic, liposome-mediated administration of genes coding for antiangiogenic factors represents a promising strategy for melanoma treatment in humans.


Subject(s)
Genetic Therapy/methods , Melanoma/therapy , Peptide Fragments/genetics , Plasminogen/genetics , Angiostatins , Animals , Cell Division , DNA/metabolism , DNA, Complementary/metabolism , Humans , Lac Operon/genetics , Liposomes/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic , Peptide Fragments/biosynthesis , Plasminogen/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Cells, Cultured
9.
Gene Ther ; 8(1): 67-70, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11402303

ABSTRACT

Tumor growth and metastasis are angiogenesis-dependent. The possibility of inhibiting tumor growth by interfering with the formation of new vessels has recently raised considerable interest. We previously reported that it is possible to inhibit primary tumor growth and metastasis in a transgenic model of spontaneous breast tumor, which shows many similarities to its human counterpart (including ability to metastasize) by intratumoral administration of a DNA construct carrying the murine angiostatin cDNA driven by liposomes. Here we report that it is also possible to achieve this goal by a systemic (intraperitoneal) delivery of therapeutic DNA constructs carrying genes coding for mouse and human anti-angiogenic factors which include angiostatin, endostatin and TIMP-2. These findings may be relevant to the design of therapeutic interventions in humans.


Subject(s)
Adenocarcinoma/therapy , Angiogenesis Inhibitors/genetics , Genetic Therapy/methods , Mammary Neoplasms, Experimental/therapy , Neovascularization, Pathologic/prevention & control , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Animals , Female , Humans , Liposomes , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic
10.
Blood ; 97(1): 81-8, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11133745

ABSTRACT

Severe combined immunodeficiency (SCID) comprises a heterogeneous group of primary immunodeficiencies, a proportion of which are due to mutations in either of the 2 recombination activating genes (RAG)-1 and -2, which mediate the process of V(D)J recombination leading to the assembly of antigen receptor genes. It is reported here that the clinical and immunologic phenotypes of patients bearing mutations in RAGs are more diverse than previously thought and that this variability is related, in part, to the specific type of RAG mutation. By analyzing 44 such patients from 41 families, the following conclusions were reached: (1) null mutations on both alleles lead to the T-B-SCID phenotype; (2) patients manifesting classic Omenn syndrome (OS) have missense mutations on at least one allele and maintain partial V(D)J recombination activity, which accounts for the generation of residual, oligoclonal T-lymphocytes; (3) in a third group of patients, findings were only partially compatible with OS, and these patients, who also carried at least one missense mutation, may be considered to have atypical SCID/OS; (4) patients with engraftment of maternal T cells as a complication of a transplacental transfusion represented a fourth group, and these patients, who often presented with a clinical phenotype mimicking OS, may be observed regardless of the type of RAG gene mutation. Analysis of the RAG genes by direct sequencing is an effective way to provide accurate diagnosis of RAG-deficient as opposed to RAG-independent V(D)J recombination defects, a distinction that cannot be made based on clinical and immunologic phenotype alone.


Subject(s)
Genes, RAG-1/genetics , Immunoglobulin Joining Region/genetics , Immunoglobulin Variable Region/genetics , Lymphocytes/immunology , Alleles , Cohort Studies , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Databases, Factual , Family Health , Female , Genotype , Humans , Immunophenotyping , Infant , Infant, Newborn , Lymphopenia/etiology , Male , Maternal-Fetal Exchange/immunology , Mutation , Mutation, Missense , Nuclear Proteins , Pregnancy , Recombination, Genetic , Severe Combined Immunodeficiency/complications , Severe Combined Immunodeficiency/genetics , T-Lymphocytes/transplantation
11.
Cancer Gene Ther ; 7(10): 1299-306, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11059686

ABSTRACT

Mutations in the CD40 ligand (CD40L) are responsible for human hyper immunoglobulin M (IgM) syndrome. The absence of the interaction between CD40L, expressed by T lymphocytes, and the CD40 receptor present on the surface of B cells is responsible for the inability of B cells to carry out the isotype switch from IgM to the other Ig classes. This leads to a fatal immunodeficiency for which no cure exists. For these reasons, the CD40L gene is a good candidate for gene therapy studies. To investigate the possible effects of the expression of this tightly regulated gene in vivo, we produced transgenic mice in which CD40L expression was deregulated. Widespread ectopic expression appears to be lethal. Overexpression in mature T cells is compatible with life, but in one-third of the cases, mice developed atypical lymphoid proliferations which, occasionally, progressed into frank lymphomas. Even though gene therapy is one of the most promising approaches to cure human hyper IgM syndrome, these results suggest that when we modify very tightly regulated genes such as cytokines or other growth factors, particular care has to be taken to avoid excessive stimulation of the target cells.


Subject(s)
CD40 Ligand/genetics , Genetic Therapy/methods , Hypergammaglobulinemia/therapy , Immunoglobulin M , Lymphoma, B-Cell/etiology , Animals , B-Lymphocytes/immunology , Blotting, Northern , CD40 Antigens/genetics , Cells, Cultured , DNA Primers/chemistry , Flow Cytometry , Gene Expression , Humans , Hypergammaglobulinemia/immunology , Immunoenzyme Techniques , Lymph Nodes/ultrastructure , Lymphocyte Activation , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Transgenic , Polymerase Chain Reaction , T-Lymphocytes/immunology
12.
Cancer Res ; 60(10): 2660-5, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10825138

ABSTRACT

The possibility to inhibit tumor growth by interfering with the formation of new vessels, which most neoplasias depend on, has recently raised considerable interest. An angiogenic switch, in which proliferating cells acquire the ability to direct new vessel formation, is thought to be an early step in the natural history of solid tumors. Using a transgenic model of breast cancer, which shows many similarities to its human counterpart, including ability to metastasize, we targeted angiostatin production to an early stage of tumor formation. Liposome-delivered angiostatin considerably delayed primary tumor growth and, more importantly, inhibited the appearance of lung metastases. These findings can be relevant to the design of therapeutic intervention in humans.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Liposomes , Mammary Neoplasms, Experimental/drug therapy , Neoplasm Metastasis/prevention & control , Peptide Fragments/administration & dosage , Peptide Fragments/therapeutic use , Plasminogen/administration & dosage , Plasminogen/therapeutic use , Angiostatins , Animals , Female , Genetic Therapy , Humans , Mammary Neoplasms, Experimental/pathology , Membrane Proteins/genetics , Mice , Mice, Transgenic , Receptor, ErbB-2/genetics , Receptors, Virus/genetics
13.
J Toxicol Environ Health A ; 55(2): 121-31, 1998 Sep 25.
Article in English | MEDLINE | ID: mdl-9761132

ABSTRACT

The effects of two substituted polychlorinated biphenyls, the 3,4,5,3',4,5' (PCB-169) and the 2,3,4,2',4',5' (PCB-138) forms, were examined on the expression of c-myc, c-jun, c-ras, and jun-b in 3T3-L1 cells. Northern blot analysis demonstrated that the two PCBs, which exhibit a coplanar and di-ortho-substituted configuration, activated these oncogenes differently. PCB-138 markedly induced overexpression of ras, jun, and myc, whereas PCB-169 led to the overexpression of jun-b. High-performance liquid chromatography analysis of the cell samples treated in medium without serum revealed a higher intracellular concentration of the 2,3,4,2',4',5'-hexachlorobiphenyl (hexaCB), whereas the 3,4,5,3',4'5'-hexaCB reached the same concentration in the sonicated samples of cells with or without serum. These results indicated that there was a relationship between PCB structure, bioavailability, and the capacity to stimulate oncogene expression.


Subject(s)
Polychlorinated Biphenyls/toxicity , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , ras Proteins/biosynthesis , 3T3 Cells , Animals , Blotting, Northern , Cell Division/drug effects , DNA Probes , Mice , Polychlorinated Biphenyls/analysis , RNA/analysis , RNA/isolation & purification
14.
Gene Ther ; 5(3): 388-93, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9614559

ABSTRACT

Oncogene-bearing transgenic mice develop various kinds of tumors depending on both the regulatory sequences and the specific oncogene used. These mice not only help to clarify the pathogenetic pathways leading to tumor formation, but can also be useful as models to test novel therapeutic strategies, including gene therapy. We have previously reported the establishment of an MMTV-neu (ErbB-2) transgenic mouse lineage, in which 100% of females develop breast tumors with many features similar to their human counterparts; these tumors are due to the over-expression of the activated rat neu oncogene in the mammary gland. From one such mouse we established a cell line of mammary adenocarcinoma named MG1361. We report here that the growth of this cell line can be inhibited in vitro and in vivo by transfection of a plasmid vector carrying an antisense anti-neu construct. This inhibitory effect is specific, as it is related to the expression of the antisense transgene (determined by RT-PCR), and to a reduction in neu mRNA and protein, as determined by Northern and Western blot analyses. Moreover, inoculation of cells carrying the antisense or the control vector in nude mice demonstrated that the morphological and biochemical effects elicited by the antisense construct resulted in a significantly slower rate of in vivo growth of tumor xenografts. Finally, significant mammary tumor growth inhibition was obtained after liposome-mediated direct inoculation of the same antisense vector in tumors spontaneously arising in MMTV-neu mice. Taken together, these findings suggest that targeting neu expression by an integrated large anti-neu antisense segment affects the in vivo growth of these tumors.


Subject(s)
Adenocarcinoma/genetics , Antisense Elements (Genetics) , Genes, erbB-2 , Mammary Neoplasms, Experimental/genetics , Adenocarcinoma/pathology , Animals , Female , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Genetic Vectors , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Transplantation , Polymerase Chain Reaction , RNA, Messenger/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Tumor Cells, Cultured
15.
Breast Cancer Res Treat ; 47(2): 171-80, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9497105

ABSTRACT

A new murine cell line, named MG1361, was established from mammary adenocarcinomas arising in a MMTV-neu transgenic mouse lineage where breast tumors develop in 100% of females, due to the overexpression of the activated rat neu oncogene in the mammary gland. The MG1361 cell line shows an epithelial-like morphology, has a poor plating efficiency, low clonogenic capacity, and a doubling time of 23.8 hours. Karyotype and flow cytometry analysis revealed a hypotetraploid number of chromosomes, whereas cell cycle analysis showed 31.2% of cells to be in the G1 phase, 21.4% in S and 47.4% in G2 + M. This cell line maintains a high level of neu expression in vitro. The MG1361 cell line was tumorigenic when inoculated in immunodeficient (nude) mice and the derived tumors showed the same histological features as the primary tumors from which they were isolated. MG1361 cells were positive for specific ER and PgR binding which was competed by tamoxifen, making this cell line useful for the evaluation of endocrine therapy. Moreover, they were sensitive to etoposide treatment, suggesting that they could be a model for the study of chemotherapy-induced apoptosis. As the tumors arising in MMTV-neu transgenic mice have many features in common with human mammary adenocarcinomas (Sacco et al., Gene Therapy 1995; 2: 493-497), this cell line can be utilized to perform basic studies on the role of the neu oncogene in the maintenance of the transformed phenotype, and to test novel protocols of therapeutic strategies.


Subject(s)
Adenocarcinoma , Breast Neoplasms , Animals , Culture Techniques , Female , Flow Cytometry , Karyotyping , Mice , Mice, Transgenic , Receptor, ErbB-2/genetics , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Tumor Cells, Cultured/ultrastructure
16.
Cancer Biochem Biophys ; 16(3): 229-42, 1998 Oct.
Article in English | MEDLINE | ID: mdl-10072207

ABSTRACT

Several studies have demonstrated that transfer of oncogenes in cultured cells reproducibly induces transmissible alterations in their ganglioside profile; the transfection of the same oncogene into different cell lines and the different localization of the oncogene product result in a different ganglioside expression. In the present study the modifications of the ganglioside pattern in mammary carcinomas induced in transgenic mice by the activated form of the rat neu oncogene have been investigated. Whereas control mammary tissues contain quite exclusively GM3, all neoplastic samples show a substantial decrease of this ganglioside, an accumulation in variable amount of GM3-derived species (GM1, GD3, GD1a, GD1b, GT and GQ) and the appearance of new, not yet identified, sialic acid containing molecules. Interestingly, three out of 10 tumors analyzed, even if histologically comparable to the others but with a larger dimension, show a significative difference as regard to the GM1, GD3 and GD1a content. Our data suggest that an activated oncogene may induce also in vivo a specific and transmissible alteration in the ganglioside pattern, but this distribution could be susceptible to further modifications during the tumor progression.


Subject(s)
Gangliosides/metabolism , Genetic Engineering/methods , Mice, Transgenic , Oncogenes , Animals , Chromatography, Thin Layer , Female , Gangliosides/analysis , Genes, erbB-2 , Hydrogen-Ion Concentration , Male , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/metabolism , Mice , N-Acetylneuraminic Acid/metabolism , Proto-Oncogene Proteins/metabolism
17.
Breast Cancer Res Treat ; 45(2): 141-7, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9342439

ABSTRACT

The Insulin Receptor (IR) is a potential oncogene for mammary epithelial cells since its content is increased in most human breast cancer specimens, and both ligand-dependent malignant transformation and ligand-dependent enhanced growth occurs in cultured breast cells overexpressing the IR. To better understand whether the IR plays a role in mammary carcinogenesis which is independent of other initiation factors, we measured IR content in transgenic mouse models of breast cancer induced by 3 known oncogenes (Wnt-1, Neu, and Ret). Insulin receptor content was measured by a specific radioimmunoassay. In normal mammary gland tissues IR content was 14.6 +/- 1.4 ng/mg of protein (mean +/- SEM, n = 6). In the 3 cancers IR content was elevated (Neu = 36.1 +/- 4.6, n = 8, p < 0.002; Wnt-1 = 38.3 +/- 2.6, n = 13, p < 0.001; and Ret = 53.6 +/- 7.1, n = 7, p < 0.001). These data indicate that IR overexpression, in addition to being a potential oncogene, is increased in mouse tumors initiated by other oncogenes, and therefore may also play a supportive role in the growth of breast cancers.


Subject(s)
Mammary Neoplasms, Animal/chemistry , Oncogenes/genetics , Receptor, Insulin/analysis , Animals , Female , Male , Mammary Glands, Animal/chemistry , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Radioimmunoassay
18.
Nat Biotechnol ; 15(13): 1392-7, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9415893

ABSTRACT

Transgenic mice for genotoxicity testing have been developed, although no such models have been produced for the evaluation of toxic, nongenotoxic chemical compounds. We have developed a transgenic mouse model for the analysis of toxic inorganic compounds. We engineered a mouse lineage with the human growth hormone (hGH) gene under the control of the human hsp70 promoter, in which a plasma-detectable hGH response can be elicited by exposure to heat shock. In primary cell cultures from these mice, hGH release was observed following treatment with several toxic inorganics. Transgenic mice injected intraperitoneally with sodium arsenite, cadmium chloride, copper sulphate, or methylmercurium chloride showed significant hGH levels in plasma.


Subject(s)
HSP70 Heat-Shock Proteins/genetics , Human Growth Hormone/genetics , Liver/drug effects , Mutagenicity Tests , Xenobiotics/toxicity , Animals , Arsenites/administration & dosage , Arsenites/toxicity , Cadmium Chloride/administration & dosage , Cadmium Chloride/toxicity , Cells, Cultured/drug effects , Copper Sulfate/administration & dosage , Copper Sulfate/toxicity , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HSP70 Heat-Shock Proteins/blood , Human Growth Hormone/biosynthesis , Human Growth Hormone/blood , Humans , Injections, Intraperitoneal , Liver/metabolism , Methylmercury Compounds/administration & dosage , Methylmercury Compounds/toxicity , Mice , Mice, Transgenic , Models, Genetic , Polymerase Chain Reaction , Promoter Regions, Genetic , Sodium Compounds/administration & dosage , Sodium Compounds/toxicity , Transgenes
19.
Gene Ther ; 3(12): 1151-6, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8986442

ABSTRACT

Mice transgenic for the activated rat neu oncogene under the control of the mouse mammary tumor virus long terminal repeat (MMTV-LTR) (neu+ mice), develop breast tumors in 100% of cases. We have previously reported that double transgenic mice obtained from crossing neu+ mice with mice transgenic for the herpes simplex virus thymidine kinase (HSVtk) gene can be used as a suitable model to test the 'suicide gene' strategy for mammary tumor gene therapy in vivo. In the present study, we evaluated the efficacy of the HSVtk/ganciclovir (GCV) system in the neu+ mice by inoculating cells producing a retroviral vector bearing the HSVtk gene in the mammary tumors on one side of the animals, and comparing their weight with that of the contralateral tumors, after systemic GCV administration. A statistically significant effect of this therapy was clearly seen (P < 0.001) but complete eradication of the tumors could not be achieved. This was not due to the inefficient delivery of GCV, as no HSVtk expression was detected in the residual tumors, but could be related to the low transduction efficiency (< 10%) and to inability of the 'bystander effect' (probably due to the absence of functional gap-junctions among mammary tumor cells) to kill nontransduced neoplastic cells. These data suggest that results obtained by in vivo models using transplanted tumor cell lines as targets for gene therapy might not be immediately transferable to spontaneously arising tumors in animals or humans.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents/therapeutic use , Ganciclovir/therapeutic use , Gene Transfer Techniques , Genetic Therapy , Mammary Neoplasms, Experimental/therapy , Mammary Tumor Virus, Mouse/genetics , Thymidine Kinase/genetics , Adenocarcinoma/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line/transplantation , Female , Ganciclovir/pharmacokinetics , Genes, erbB-2 , Genetic Vectors , Humans , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Transgenic , Rats , Thymidine Kinase/metabolism , Tumor Cells, Cultured
20.
Genomics ; 32(2): 277-80, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8833156

ABSTRACT

The gene encoding the human host cell factor (HCFC1) has recently been cloned and mapped to Xq28. HCFC1 codes for a family of related polypeptides that apparently arise from posttranslational processing. Six extremely conserved 19-amino-acid (aa)long motifs, unique to HCFC1 and located in the middle of the protein, could play a role in this processing or could be instrumental to the physiological role of the protein. Aternatively, these repeats could have arisen from recent duplications and may not have any specific function. To resolve this issue, we cloned the homologous region from the mouse HCFC1 gene and demonstrated that the 19-aa motifs are extremely conserved in sequence, number, and genomic organization, while the "linker" region between the third and fourth repeat is not. This suggests an important function for these repeats. In addition, by RT-PCR analysis of human RNA and comparison to the human genomic sequence, an alternative transcript including a 44-aa in-frame insertion, deriving from the 3' end of intron 18, was found. The significance of this alternative transcript is unknown, since it was not detectable in the mouse. The mouse HCFC1 gene maps to a region syntenic to Xq28, and, as in human, is in close proximity to the Renin-binding protein gene, in a 100-kb region also including the Licam and Vasopressin receptor type 2 genes.


Subject(s)
Conserved Sequence , Proteins/genetics , Transcription Factors , X Chromosome , Amino Acid Sequence , Animals , Chromosome Mapping , Chromosomes, Artificial, Yeast , Host Cell Factor C1 , Humans , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...