Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 388
Filter
1.
Regul Toxicol Pharmacol ; 50(1): 87-97, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18069108

ABSTRACT

The use of transgenic rodents may overcome many limitations of traditional cancer studies. Regulatory perspectives continue to evolve as new models are developed and validated. The transgenic mouse, K6/ODC, develops epidermal tumors when exposed to genotoxic carcinogens. In this study, K6/ODC mice were evaluated for model fitness and health robustness in a 36-week study to determine oncogenic risk of residual DNA in vaccines from neoplastic cell substrates. K6/ODC and C57BL/6 mice were treated with T24-H-ras expression plasmid, carrier vector DNA, or saline topically or by subcutaneous injection. One group of K6/ODC mice received 7,12-dimethylbenz-[a]anthracene [DMBA] dermally. Only DMBA-treated mice developed papillomas by six weeks, increasing in incidence to 25 weeks. By week 11, many K6/ODC mice showed severe dehydration and dermal eczema. By week 32, (6/8) surviving K6/ODC mice showed loss of mobility and balance. Microscopic evaluation of tissues revealed dermal/sebaceous gland hyperplasia, follicular dystrophy, splenic atrophy, and amyloid deposition/neutrophilic infiltration within liver, heart, and spleen, in all K6/ODC mice. Pathology was not detected in C57BL/6 mice. Progressive adverse health, decreased survival, and failure to develop papillomas to the H-ras plasmid suggest that K6/ODC mice may be an inappropriate alternative model for detection of oncogenic DNA and pharmaceutical carcinogenicity testing.


Subject(s)
Disease Models, Animal , Drug Evaluation, Preclinical/methods , Keratin-6/genetics , Ornithine Decarboxylase/genetics , Skin Neoplasms/chemically induced , 9,10-Dimethyl-1,2-benzanthracene/administration & dosage , Animals , Carcinogenicity Tests/methods , Carcinogens/administration & dosage , Drug-Related Side Effects and Adverse Reactions , Kidney/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sarcoma Viruses, Murine/genetics , Skin Neoplasms/pathology , Spleen/drug effects , Spleen/pathology , Transfection
2.
Biochem Biophys Res Commun ; 343(4): 1017-22, 2006 May 19.
Article in English | MEDLINE | ID: mdl-16574062

ABSTRACT

The long terminal repeat (LTR) of retrovirus contains the nucleotide sequences that control gene expression. Although several different LTRs have been used in the context of retroviral vector, the activity of the various LTRs has not yet been systematically compared for their level of gene expression. We evaluated the effect of four different LTRs on gene expression using luciferase, stem cell factor, and enhanced green fluorescence protein as reporter genes. LTRs tested in this study were derived from Moloney murine leukemia virus, myeloproliferative sarcoma virus, murine stem cell virus, and spleen focus-forming virus. It was found that the level of gene expression is affected by not only LTRs but also the transgenes and the cell types in which gene expression occurs. Furthermore, the presence of other nucleotide sequences such as the internal ribosome entry site (IRES)-neo cassette could also significantly affect gene expression. Our results suggested that the LTR should be chosen carefully, more or less on an empirical basis.


Subject(s)
Gene Expression Regulation , Genetic Vectors , Retroviridae/genetics , Terminal Repeat Sequences , Transgenes , Cell Line, Tumor , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Leukemia Virus, Murine/genetics , Luciferases/biosynthesis , Luciferases/genetics , Moloney murine leukemia virus/genetics , Sarcoma Viruses, Murine/genetics , Stem Cell Factor/biosynthesis , Stem Cell Factor/genetics
3.
Protein Expr Purif ; 45(1): 115-24, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16289982

ABSTRACT

With the recent completion of the human genome sequencing project, scientists are faced with the daunting challenge of deciphering the function of these newly found genes quickly and efficiently. Equally as important is to produce milligram quantities of the therapeutically relevant gene products as quickly as possible. Mammalian expression systems provide many advantages to aid in this task. Mammalian cell lines have the capacity for proper post-translational modifications including proper protein folding and glycosylation. In response to the needs described above, we investigated the protein expression levels driven by the human CMV in the presence or absence of intron A, the mouse and rat CMV promoters with intron A, and the MPSV promoter in plasmid expression vectors. We evaluated the different promoters using an in-house plasmid vector backbone. The protein expression levels of four genes of interest driven by these promoters were evaluated in HEK293EBNA and CHO-K1 cells. Stable and transient transfected cells were utilized. In general, the full-length human CMV, in the presence of intron A, gave the highest levels of protein expression in transient transfections in both cell lines. However, the MPSV promoter resulted in the highest levels of stable protein expression in CHO-K1 cells. Using the CMV driven constitutive promoters in the presence of intron A, we have been able to generate >10 microg/ml of recombinant protein using transient transfections.


Subject(s)
Cytomegalovirus/genetics , Gene Expression Regulation , Genetic Vectors/genetics , Promoter Regions, Genetic , Animals , CHO Cells , Cell Line , Cricetinae , Humans , Mice , RNA, Messenger/genetics , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Sarcoma Viruses, Murine/genetics , Transcription, Genetic/genetics , Transfection
4.
Ukr Biokhim Zh (1999) ; 77(2): 5-15, 2005.
Article in English | MEDLINE | ID: mdl-16335231

ABSTRACT

Genome of all known retroviruses consists of two identical molecules of RNA, which are non-covalently linked. The most stable contact site between two RNA molecules is located near their 5' ends. The molecular interactions in the dimer linkage structure (DLS) in mature virions are currently unknown. Recently we suggested that the dimer linkage structure in human immunodeficiency virus 1 (HIV-1) contains both duplex and quadruplex domains and proposed a model of DLS in HIV-1Mal (Central African virus). In this paper we showed that similar models can be also built for HIV- 1Lai, a representative of the North-American and European viruses. One of the double-stranded domains in the model structures represents either an extended duplex formed by different pathways (through base pair melting and subsequent reannealing or by a recombination mechanism) or kissing loop complex. The quadruplexes contain both G- and mixed tetrads, for example, G.C.G.C or A.U.A.U. Phylogenetic analysis of 350 isolates from NCBI database showed that similar models of DLS are predictable practically for all HIV-1 isolates surveyed. A model of dimer linkage structure in Moloney murine sarcoma virus (MuSV) is also presented. The structure includes a duplex formed by the palindromic sequences and several quadruplexes.


Subject(s)
Genome, Viral , RNA, Viral/chemistry , Retroviridae/genetics , Animals , Dimerization , HIV-1/genetics , Humans , Mice , Models, Biological , RNA, Viral/genetics , Sarcoma Viruses, Murine/genetics
5.
Hum Gene Ther ; 16(7): 799-810, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16000062

ABSTRACT

Adoptive T cell therapy of renal cell carcinoma (RCC) is limited by the difficulty in generating sufficient numbers of RCC-reactive T cells in vitro. To circumvent this problem, we cloned T cell receptor (TCR) alpha and beta chains from a tumor-infiltrating lymphocyte clone specific for an RCC tumor antigen and transferred the TCR into human T cell lines and primary T lymphocytes. Efficient TCR expression in primary T lymphocytes was obtained only with a mouse myeloproliferative sarcoma virus (MPSV)-based retroviral vector, not with a Moloney murine leukemia virus (MLV)-based vector, although both viral supernatants were similar in titer, as shown by analysis of copy number integration in transduced T cells. Reverse transcription-polymerase chain reaction analysis revealed a higher amount of TCR-encoding transcripts when T cells were transduced with the MPSV vector in comparison with the MLV vector, indicating that high TCR expression levels can be achieved by appropriate cis-regulatory vector elements. The biological activity of the transferred TCR was shown by specific lysis of RCC cells ((51)Cr release assay) and by interferon gamma and tumor necrosis factor alpha release (enzyme-linked immunosorbent assay) in an antigen-specific and HLA-A*0201-restricted fashion. Comparison of the redirected T lymphocytes with the original tumor-infiltrating lymphocyte clone revealed similar killing and cytokine secretion capabilities. The functional activity of TCR-redirected T lymphocytes was stable over time. The results demonstrate that use of an optimized retroviral vector yielded a high TCR transduction efficiency and stable and high TCR expression in primary human T lymphocytes and redirected their specificity toward RCC cells.


Subject(s)
Carcinoma, Renal Cell/immunology , Genes, T-Cell Receptor , Kidney Neoplasms/immunology , Retroviridae/genetics , T-Lymphocytes, Cytotoxic/immunology , Cell Line , Cells, Cultured , Gene Expression , Genetic Vectors , Humans , Interferon-gamma/metabolism , Moloney murine leukemia virus/genetics , Recombination, Genetic , Sarcoma Viruses, Murine/genetics , Transduction, Genetic , Tumor Necrosis Factor-alpha/metabolism
6.
J Virol ; 76(9): 4643-8, 2002 May.
Article in English | MEDLINE | ID: mdl-11932430

ABSTRACT

We investigated the features of the Moloney murine sarcoma virus leader sequence necessary for RNA packaging function by using a deletion analysis approach. We found that sequences that extend beyond those characterized genetically in previous reports are important for optimal packaging efficiency. A fragment covering a minimum of four potential stem-loop structures is required for the shortest packaging element compatible with gene transfer. Our results reveal the extent to which each of the segments of the packaging sequence contribute to packaging efficiency.


Subject(s)
5' Untranslated Regions , RNA, Viral/metabolism , Sarcoma Viruses, Murine/metabolism , Virus Assembly , Animals , Gene Transfer Techniques , Mice , Molecular Sequence Data , RNA, Viral/genetics , Sarcoma Viruses, Murine/genetics , Sequence Deletion
7.
Eur J Haematol ; 67(1): 2-13, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11553261

ABSTRACT

Overexpression of O(6)-methylguanine DNA methyltransferase (MGMT) can protect hematopoietic cells from O(6)-alkylation damage. To identify possible clinical applications of this technology we compared the effect of MGMT gene transfer on the hematotoxicity induced by different O(6)-alkylating agents in clinical use: the chloroethylnitrosoureas ACNU, BCNU, CCNU and the tetrazine derivative temozolomide. In addition, various retroviral vectors expressing the MGMT-cDNA were investigated to identify optimal viral backbones for hematoprotection by MGMT expression. Protection from ACNU, BCNU, CCNU or temozolomide toxicity was evaluated utilizing a Moloney murine leukemia virus-based retroviral vector (N2/Zip-PGK-MGMT) to transduce primary murine bone marrow cells. Increased resistance in murine colony-forming units (CFU) was demonstrated for all four drugs. In comparison to mock-transduced controls, after transduction with N2/Zip-PGK-MGMT the IC50 for CFU increased on average 4.7-fold for ACNU, 2.5-fold for BCNU, 6.3-fold for CCNU and 1.5-fold for temozolomide. To study the effect of the retroviral backbone on hematoprotection various vectors expressing the human MGMT-cDNA from a murine embryonic sarcoma virus LTR (MSCV-MGMT) or a hybrid spleen focus-forming/murine embryonic sarcoma virus LTR (SF1-MGMT) were compared with the N2/Zip-PGK-MGMT vector. While all vectors increased resistance of transduced human CFU to ACNU, the SF1-MGMT construct was most efficient especially at high ACNU concentrations (8-12 microg/ml). Similar results were obtained for protection of murine high-proliferative-potential colony-forming cells. These data may help to optimize treatment design and retroviral constructs in future clinical studies aiming at hematoprotection by MGMT gene transfer.


Subject(s)
Alkylating Agents/toxicity , DNA Damage , Drug Resistance/genetics , Genetic Vectors/genetics , Hematopoietic Stem Cells/drug effects , O(6)-Methylguanine-DNA Methyltransferase/physiology , Retroviridae/genetics , Alkylation , Animals , Carmustine/toxicity , Cells, Cultured , Colony-Forming Units Assay , DNA, Complementary/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/toxicity , Hematopoietic Stem Cells/enzymology , Humans , Lomustine/toxicity , Mice , Moloney murine leukemia virus/genetics , Nimustine/toxicity , O(6)-Methylguanine-DNA Methyltransferase/genetics , Recombinant Fusion Proteins/physiology , Sarcoma Viruses, Murine/genetics , Spleen Focus-Forming Viruses/genetics , Temozolomide , Terminal Repeat Sequences , Transfection
8.
Oncogene ; 20(8): 942-50, 2001 Feb 22.
Article in English | MEDLINE | ID: mdl-11314029

ABSTRACT

c-Fos proto-oncoprotein is a short-lived transcription factor degraded by the proteasome in vivo. Its mutated forms expressed by the mouse osteosarcomatogenic retroviruses, FBJ-MSV and FBR-MSV, are stabilized two- and threefold, respectively. To elucidate the mechanisms underlying v-Fos(FBJ) and v-Fos(FBR) protein stabilization, we conducted a genetic analysis in which the half-lives and the sensitivities to various cell-permeable protease inhibitors of a variety of cellular and viral protein mutants were measured. Our data showed that the decreased degradation of v-Fos(FBJ) and v-Fos(FBR) is not simply explained by the deletion of a c-Fos destabilizing C-terminal domain. Rather, it involves a complex balance between opposing destabilizing and stabilizing mutations which are distinct and which include virally-introduced peptide motifs in both cases. The mutations in viral Fos proteins conferred both total insensitivity to proteasomal degradation and sensitivity to another proteolytic system not naturally operating on c-Fos, explaining the limited stabilization of the two proteins. This observation is consistent with the idea that FBR-MSV and FBJ-MSV expression machineries have evolved to ensure controlled protein levels. Importantly, our data illustrate that the degradation of unstable proteins does not necessarily involve the proteasome and provide support to the notion that highly related proteins can be broken down by different proteolytic systems in living cells.


Subject(s)
Oncogene Proteins v-fos/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Amino Acid Sequence , Animals , COS Cells , Cysteine Endopeptidases/metabolism , Frameshift Mutation , Half-Life , Multienzyme Complexes/metabolism , Myristic Acid/metabolism , Oncogene Proteins v-fos/genetics , Point Mutation , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sarcoma Viruses, Murine/genetics , Sequence Deletion
9.
Eur J Cell Biol ; 79(9): 583-93, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11043399

ABSTRACT

We have analysed the expression of cadherin/catenin complex molecules in PC C13 rat thyroid cells transformed in vitro with different oncogenes. No significant downregulation of either E-cadherin, alpha-, beta- and gamma-catenin was detected following the introduction of activated forms of myc, adenovirus E1A, ras, raf, myc + ras, E1A + raf. However, ras- and raf-transformed PC C13 cells showed altered adherens junctions. An altered distribution of cadherin/catenin complexes characterized by radially oriented membrane spikes perpendicular to cell edges was the most prominent feature evidenced by immunofluorescence. No beta1 integrin localization was observed in areas where this altered pattern of E-cadherin expression was detected. However, beta1 integrin subunit expression was detected at areas of cell-cell contact where E-cadherin showed a normal pattern of expression. Furthermore, ras- and raf-transformed PC C13 cells showed the ability to migrate in collagen gels, in contrast to their normal untransformed counterpart. Overexpression of beta1 integrin was found to restore normal E-cadherin localization at cell-cell contacts and to partially inhibit the ability to migrate in collagen gels. Finally, two cell lines obtained by ras transformation in vivo, and derived from a rat primary thyroid carcinoma (TK6) and its lung metastasis (MPTK6), were found to have lost gamma-catenin expression. TK6 lost also E-cadherin expression and membrane localization of alpha-catenin. These results suggest that: i) in vitro thyroid cell transformation is associated to a change in cadherin/catenin complexes distribution rather than to a decrease in expression; ii) in vivo transformation is associated to the loss of expression of some of these molecules likely due to tumor progression; iii) alterations in beta1 integrin subunit expression can result in changes in cadherin/catenin function thus implying that an integrin-cadherin synergy may exist in thyroid cells.


Subject(s)
Cadherins/metabolism , Cytoskeletal Proteins/metabolism , Epithelial Cells/metabolism , Integrin beta1/metabolism , Thyroid Gland/cytology , Trans-Activators , Adenovirus E1A Proteins/genetics , Animals , Blotting, Western , Cadherins/analysis , Cadherins/genetics , Cell Communication/physiology , Cell Line, Transformed , Cell Movement/physiology , Collagen , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/genetics , Desmoplakins , Epithelial Cells/chemistry , Epithelial Cells/cytology , Fluorescent Antibody Technique , Gels , Gene Expression/physiology , Genes, myc , Genes, ras , Integrin beta1/analysis , Integrin beta1/genetics , Oncogene Proteins v-raf , Rats , Retroviridae Proteins, Oncogenic/genetics , Sarcoma Viruses, Murine/genetics , alpha Catenin , beta Catenin , gamma Catenin
10.
Biotechniques ; 28(4): 702-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10769748

ABSTRACT

We have developed a retroviral vector that incorporates unique features of some previously described vectors. This vector includes: 3' long terminal repeats (LTRs) of the self-inactivating class; a 5' LTR that is a hybrid of the cytomegalovirus (CMV) enhancer and the mouse sarcoma virus promoter; an internal CMV immediate early region promoter to drive expression of the transduced gene and the neomycin phosphotransferase selectable marker; an expanded multiple cloning site and an internal ribosome entry site. An SV40 ori was introduced into the vector backbone to promote high copy number replication in packaging cell lines that express the SV40 large T antigen. We demonstrate that these retroviral constructs, designated Q vectors, can be used in applications where high viral titers and high level stable or transient gene expression are desirable.


Subject(s)
Gene Transfer Techniques , Genes/genetics , Genetic Vectors/genetics , Retroviridae/genetics , Zebrafish Proteins , Animals , Antigens, Viral, Tumor/metabolism , Cell Line , Cytomegalovirus/genetics , Epithelial Cells/cytology , Epithelial Cells/virology , Female , Fibroblasts/cytology , Fibroblasts/virology , Genes, Reporter/genetics , Genetic Vectors/chemistry , Mammary Glands, Animal/cytology , Mammary Glands, Animal/virology , Mice , Plasmids/chemistry , Plasmids/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Rats , Retroviridae/growth & development , Sarcoma Viruses, Murine/genetics , Transfection , Wnt Proteins , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
11.
Gene ; 241(1): 107-16, 2000 Jan 04.
Article in English | MEDLINE | ID: mdl-10607904

ABSTRACT

The chicken c-ski gene expresses at least three alternatively spliced messages. Transgenic mice expressing proteins from cDNA corresponding to two of these messages (FB27 and FB29) under the control of a murine sarcoma virus (MSV) long terminal repeat (LTR) express the transgene in skeletal muscle and develop a muscular phenotype. Both a biologically active form of c-ski and the MSV LTR are required for the development of the muscular phenotype. The normal c-ski gene linked to two other tissue-specific promoters failed to induce muscle growth in transgenic mice, as did an inactive mutant of c-ski expressed under the control of the MSV LTR.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Hypertrophy/metabolism , Muscle, Skeletal/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Animals , Cloning, Molecular , DNA, Complementary/metabolism , Hypertrophy/pathology , Mice , Mice, Transgenic , Models, Genetic , Muscle, Skeletal/pathology , Myosin Heavy Chains/genetics , Phenotype , Ribonucleases/metabolism , Sarcoma Viruses, Murine/genetics , Terminal Repeat Sequences/genetics , Tissue Distribution
12.
J Virol ; 73(4): 2762-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10074123

ABSTRACT

Cytotoxic T lymphocytes (CTL) appear to be critical in resolving or reducing the severity of lentivirus infections. Retroviral vectors expressing the Gag/Pr or SU protein of the lentivirus equine infectious anemia virus (EIAV) were constructed and used to evaluate EIAV-specific CTL responses in horses. Three promoters, cytomegalovirus, simian virus SV40, and Moloney murine sarcoma virus (MoMSV) long terminal repeat (LTR), were used, and there was considerable variation in their ability to direct expression of Gag/Pr and SU. Vectors expressing EIAV proteins under the direction of MoMSV LTR and using the gibbon ape leukemia virus (GALV) Env for internalization were efficient at transducing equine kidney (EK) target cells and were effective targets for EIAV-specific CTL lysis. CTL from EIAV-infected horses caused lysis of retroviral vector-transduced EK cells expressing either Gag/Pr or SU in an ELA-A-restricted manner. In contrast, lysis of recombinant vaccinia virus-infected EK cells expressing Gag/Pr and SU/TM was often non-LA-A restricted. Five horses were immunized by direct intramuscular injection with a mixture of retroviral vectors expressing Gag/Pr or SU, and one responded with EIAV-specific CTL. This result indicates that retroviral vector stimulation of CTL in horses needs to be optimized, perhaps by inclusion of appropriate cytokine genes in the constructs. However, the studies demonstrated that retroviral vector-transduced target cells were very effective for in vitro dissection of EIAV-specific CTL responses.


Subject(s)
Antigens, Viral/immunology , Equine Infectious Anemia/immunology , Genetic Vectors/immunology , Infectious Anemia Virus, Equine/immunology , Sarcoma Viruses, Murine/genetics , Sarcoma Viruses, Murine/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , DNA, Recombinant/immunology , Gene Products, gag/immunology , Glycoproteins/immunology , Horses/immunology , Horses/virology , Infectious Anemia Virus, Equine/genetics , Lymphocyte Activation , Viral Envelope Proteins/immunology , Viral Vaccines
13.
J Leukoc Biol ; 64(4): 441-50, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9766624

ABSTRACT

Malignant histiocytosis sarcoma virus (MHSV) arose as a recombinant of c-Harvey-ras murine sarcoma virus (Ha-MuSV) and Friend mink cell focus-forming virus (F-MCFV). It is a defective acute transforming retrovirus that, along with Friend murine leukemia helper virus (F-MuLV), induces malignant histiocytosis (MH) in susceptible adult mice. We have assessed the in vivo susceptibility to MHSV in inbred homozygous, F1 hybrid, congenic, and recombinant inbred (RI) mice. We have shown that: (1) in vivo resistance to MHSV is multigenic, regulated by MHC and non-MHC genes in a different fashion than with F-MCFV, F-MuLV, or Ha-MuSV; (2) using BXD RI mice, the resistance phenotype is linked with 95.8% probability to two linked loci, Pmv-9 and Iapls3-14, on chromosome 13 (homologous to the area of human chromosome 5 for which a chromosomal break point at position 5q35 is associated with human MH); and (3) CD4+ T cells are critical for MHSV resistance.


Subject(s)
Chromosome Mapping , Histiocytic Sarcoma/genetics , Histiocytic Sarcoma/virology , Major Histocompatibility Complex , Retroviridae Infections/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Chromosome Aberrations , Chromosomes, Human, Pair 5 , Crosses, Genetic , Female , Friend murine leukemia virus/genetics , Friend murine leukemia virus/pathogenicity , Genetic Linkage , Genetic Predisposition to Disease , Histiocytic Sarcoma/immunology , Homozygote , Humans , Immunity, Innate/genetics , Mice , Mice, Congenic , Mice, Inbred Strains , Mice, Mutant Strains , Mice, SCID , Mink Cell Focus-Inducing Viruses/genetics , Mink Cell Focus-Inducing Viruses/pathogenicity , Phenotype , Recombination, Genetic , Retroviridae Infections/genetics , Sarcoma Viruses, Murine/genetics , Sarcoma Viruses, Murine/pathogenicity
14.
Nucleic Acids Res ; 25(14): 2940-2, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9207047

ABSTRACT

We report a rapid and efficient PCR-based rescue procedure for integrated recombinant retroviruses. Full-length proviral DNA is amplified by long-range PCR using a pair of primers derived from the long terminal repeats (LTR), and virus is regenerated by transfecting retrovirus-packaging cells with the PCR-derived provirus. The viral yield from the PCR product is similar to that from the retroviral plasmid vector, and the representation of different inserts is accurately maintained in the recovered retroviral population. This procedure is suitable for expression cloning from retroviral libraries and should be applicable to the analysis of natural retrovirus populations.


Subject(s)
Genetic Vectors , Moloney murine leukemia virus/genetics , Sarcoma Viruses, Murine/genetics , Virus Integration , Cloning, Molecular , Genetic Vectors/isolation & purification , Humans , Moloney murine leukemia virus/isolation & purification , Polymerase Chain Reaction , Sarcoma Viruses, Murine/isolation & purification , Tumor Cells, Cultured
15.
Biochem J ; 323 ( Pt 3): 685-92, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9169601

ABSTRACT

The c-Fos and c-Jun transcription factors are rapidly turned over in vivo. One of the multiple pathways responsible for their breakdown is probably initiated by calpains, which are cytoplasmic calcium-dependent cysteine proteases. The c-fos gene has been transduced by two murine oncogenic retroviruses called Finkel-Biskis-Jenkins murine sarcoma virus (FBJ-MSV) and Finkel-Biskis-Reilly murine sarcoma virus (FBR-MSV); c-jun has been transduced by the chicken avian sarcoma virus 17 (ASV17) retrovirus. Using an in vitro degradation assay, we show that the mutated v-FosFBR, but not v-FosFBJ or v-JunASV17, is resistant to calpains. This property raises the interesting possibility that decreased sensitivity to calpains might contribute to the tumorigenic potential of FBR-MSV by allowing greater accumulation of the protein that it encodes in infected cells. It has also been demonstrated that resistance to cleavage by calpains does not result from mutations that have accumulated in the Fos moiety of the viral protein but rather from the addition of atypical peptide motifs at its both ends. This observation raises the interesting possibility that homologous regions in viral and cellular Fos either display slightly different conformations or are differentially accessible to interacting proteins.


Subject(s)
Avian Sarcoma Viruses/metabolism , Calpain/metabolism , Oncogene Protein p65(gag-jun)/metabolism , Oncogene Proteins v-fos/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Sarcoma Viruses, Murine/metabolism , Avian Sarcoma Viruses/genetics , Burkitt Lymphoma/pathology , Calcium/metabolism , Genes, fos , Genes, jun , Humans , Leukemia-Lymphoma, Adult T-Cell/pathology , Mutagenesis, Site-Directed , Neoplasm Proteins/metabolism , Oncogene Protein p65(gag-jun)/classification , Oncogene Proteins v-fos/classification , Sarcoma Viruses, Murine/genetics , Sequence Deletion , Substrate Specificity , Tumor Cells, Cultured
16.
J Cell Biol ; 135(2): 457-67, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8896601

ABSTRACT

Myristylation of FBR v-fos, a c-fos retroviral homologue that causes osteosarcomas in mice, determines many of its transcriptional properties in vitro. To determine whether myristylation of FBR v-fos contributes to in vivo tumorigenicity, we examined its transforming capability in comparison to a nonmyristylated FBR v-fos (G2A-R). Retroviral infections with FBR v-fos and G2A-R transform BALB/c-3T3 cells. The number, size, and cellular morphology of foci generated by both FBR and G2A-R are indistinguishable. However, marked biological differences were found in transgenic mice expressing either the myristylated FBR v-fos or the nonmyristylated G2A-R. 11 of 26 FBR v-fos transgenic mice died as a result of gross tumor burden. None of the 28 G2A-R transgenic mice died from tumor burden, and only two of the G2A-R mice developed bone tumors. Histologic examination of the tumors reveals that the FBR v-fos bone tumors contain malignant cells with features of four cell lineages (osteocytes, chondrocytes, myocytes, and adipocytes) in an environment rich in extracellular matrix (ECM). However, the G2A-R tumors exist in an environment devoid of ECM and display malignant cells with features of adipocytes. Masson staining reveals that the ECM of the FBR tumors stains strongly for collagen. Immunohistochemical staining with collagen III antibody demonstrates an abundance of collagen III expression in this ECM. While NH2-terminal myristylation is not required for FBR immortalization and transformation, it is essential in determining the degree of differentiation and tumorigenicity of malignant cells.


Subject(s)
Cell Transformation, Neoplastic , Myristic Acids/metabolism , Oncogene Proteins v-fos/metabolism , Osteosarcoma/pathology , Protein Processing, Post-Translational , Sarcoma Viruses, Murine/genetics , 3T3 Cells , Animals , Base Sequence , Cell Differentiation , Mice , Mice, Inbred BALB C , Mice, Transgenic , Moloney murine leukemia virus/genetics , Myristic Acid , Osteosarcoma/genetics , Polymerase Chain Reaction , Repetitive Sequences, Nucleic Acid
17.
Proc Natl Acad Sci U S A ; 93(15): 7522-7, 1996 Jul 23.
Article in English | MEDLINE | ID: mdl-8755507

ABSTRACT

The host range of retroviral oncogenes is naturally limited by the host range of the retroviral vector. The question of whether the transforming host range of retroviral oncogenes is also restricted by the host species has not been directly addressed. Here we have tested in avian and murine host species the transforming host range of two retroviral onc genes, myc of avian carcinoma viruses MH2 and MC29 and mht/raf of avian carcinoma virus MH2 and murine sarcoma virus MSV 3611. Virus vector-mediated host restriction was bypassed by recombining viral oncogenes with retroviral vectors that can readily infect the host to be tested. It was found that, despite high expression, transforming function of retroviral myc genes is restricted to avian cells, and that of retroviral mht/raf genes is restricted to murine cells. Since retroviral oncogenes encode the same proteins as certain cellular genes, termed protooncogenes, our data must also be relevant to the oncogene hypothesis of cancer. According to this hypothesis, cancer is caused by mutation of protooncogenes. Because protooncogenes are conserved in evolution and are presumed to have conserved functions, the oncogene hypothesis assumes no host range restriction of transforming function. For example, mutated human proto-myc is postulated to cause Burkitt lymphoma, because avian retroviruses with myc genes cause cancer in birds. But there is no evidence that known mutated protooncogenes can transform human cells. The findings reported here indicate that host range restriction appears to be one of the reasons (in addition to insufficient transcriptional activation) why known, mutated protooncogenes lack transforming function in human cells.


Subject(s)
Cell Transformation, Neoplastic , Genes, myc , Oncogenes , Retroviridae Proteins, Oncogenic/genetics , Retroviridae/genetics , Animals , Biological Evolution , Birds , Cell Line , Cells, Cultured , Chromosome Mapping , Embryo, Mammalian , Embryo, Nonmammalian , Genetic Vectors , Humans , Oncogene Proteins v-raf , Proto-Oncogenes , Rats , Rats, Inbred F344 , Sarcoma Viruses, Murine/genetics , Species Specificity , Transfection
18.
Hum Gene Ther ; 7(9): 1111-22, 1996 Jun 10.
Article in English | MEDLINE | ID: mdl-8773513

ABSTRACT

The ability to select bone marrow cells (BMC) expressing a selectable gene that confers resistance to anticancer drugs would be useful to protect bone marrow during chemotherapy. The human multidrug resistance (MDR1) gene encodes a 170-kD glycoprotein (P-gp), an ATP-dependent transmembrane efflux pump for many different cytotoxic drugs. In this work, we demonstrate efficient expression of the human MDR1 gene in mouse BMC after transfection with a liposomal delivery system (DLS-liposomes). The human MDR1 cDNA expression plasmid (pHaMDR1/A) was encapsulated in DLS-liposomes and delivered to mouse BMC using two approaches: (i) in vitro transfection of BMC followed by bone marrow transplantation and (ii) in vivo direct systemic gene transfer. After both the in vitro and the in vivo approaches, polymerase chain reaction (PCR) analysis confirmed that the human MDR1 gene was successfully transfected to bone marrow, spleen, and peripheral blood (PB) cells, with the human MDR1 gene detected in BMC for up to 30 days after bone marrow transplantation and 28 days after direct systemic administration. Efflux studies using rhodamine-123 demonstrated function of the MDR1 gene product in the in vitro-transfected BMC. Flow cytometry studies using the human MDR1-specific MRK16 monoclonal antibody confirmed the presence of P-gp in BMC after in vitro transfection, as well as in BMC from reconstituted or in vivo-transfected mice. Transgene expression in both lymphoid and myeloid subpopulations of BMC was demonstrated. Colony-forming units (CFU-Mix) were obtained after exposure of BMC to lethal doses of vincristine, demonstrating functional expression of the MDR1 gene in hematopoietic progenitor cells for up to 1 month.


Subject(s)
Gene Expression Regulation/physiology , Gene Transfer Techniques , Genes, MDR , Hematopoietic Stem Cells/metabolism , Repetitive Sequences, Nucleic Acid , Sarcoma Viruses, Murine/genetics , 3T3 Cells , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Animals , Cell Transplantation , DNA, Complementary/genetics , DNA, Viral/genetics , Drug Carriers , Humans , Liposomes , Mice , Mice, Inbred C57BL
19.
J Biol Chem ; 271(15): 9141-9, 1996 Apr 12.
Article in English | MEDLINE | ID: mdl-8621566

ABSTRACT

The MyoD family of transcription factors regulates muscle-specific gene expression in vertebrates. In the adult rat, MyoD mRNA accumulates predominately in fast-twitch muscle, in particular type IIb and/or IIx fibers, whereas Myogenin mRNA is restricted to slow-twitch type I muscle fibers. Transgenic mice expressing the avian v-ski oncogene from the murine sarcoma virus (MSV) promoter-enhancer display preferential hypertrophy of type IIb fast-twitch muscle apparently because of the restricted expression of the transgene. We tested the hypothesis that preferential interactions of MyoD, as a heterodimer with E12, with the MSV enhancer, which has six E-box targets for MyoD family proteins, could contribute to v-ski gene expression in IIb muscle fibers. A series of quantitative binding studies was performed using an electrophoretic mobility shift assay to test MyoD-E12 versus Myogenin-E12 binding to the MSV enhancer. Our results indicate that MyoD-E12 binds the MSV enhancer with higher affinity and higher cooperativity than Myogenin-E12. Interestingly, MyoD-E12 bound all of the individual E-boxes tested with positive cooperativity indicating DNA-mediated dimerization of the protein subunits.


Subject(s)
Enhancer Elements, Genetic , MyoD Protein/metabolism , Myogenin/metabolism , Sarcoma Viruses, Murine/genetics , Animals , Base Sequence , DNA/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Mice , Mice, Transgenic , Molecular Sequence Data , Retroviridae Proteins, Oncogenic/metabolism , Sequence Deletion , Structure-Activity Relationship , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein , Transcription Factors/metabolism
20.
Blood ; 87(8): 3218-28, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8605337

ABSTRACT

A strictly stroma-dependent hematopoietic clone, Myl-D-7, with lympho-myeloid potential has been isolated. A subset of cells expresses myeloid-macrophage (Mac-1 and Gr-1), erythroid (TER119), and lymphoid (Thy-1 and B220) lineage markers. Spontaneous differentiation to the myeloid-macrophage, erythroid, or lymphoid pathway can be seen by morphologic criteria, detection of beta major globin synthesis, or expression of the early lymphoid specific transcription factor, Ikaros. By sorting lineage marker (Mac-1, Gr-1, B220, and TER119)-negative (LIN-) cells, we showed that the LIN- population actively self-renews on top of MS-5 stromal cells, and differentiates to LIN+ cells. Removal of stroma induces apoptosis and none of the growth factors tested can prevent apoptosis. Granulocyte-macrophage colony-stimulating factor accelerates the differentiation towards the myeloid-macrophage lineage. Using this clone, we show that (1) contact with stroma induces expression of bcl-2, (2) stromal cells derived from SI/SI homozygous fetuses can support long-term growth, and (3) conditioned media of specific stromal cells contains an activity that supports proliferation and self-renewal of the clone. Myl-D-7 can thus be used as an indicator cell for unknown factors that may provide stromal cell support.


Subject(s)
Biological Factors/physiology , Bone Marrow Cells , Connective Tissue/physiology , Hematopoietic Stem Cells/cytology , Animals , Apoptosis , Biomarkers/analysis , Cell Differentiation/drug effects , Cell Line , Cell Lineage , Clone Cells/cytology , Coculture Techniques , Colony-Forming Units Assay , Connective Tissue Cells , Culture Media, Conditioned , Defective Viruses/genetics , Female , Friend murine leukemia virus/genetics , Genetic Vectors/genetics , Helper Viruses/genetics , Hematopoietic Cell Growth Factors/analysis , Hematopoietic Cell Growth Factors/genetics , Hematopoietic Cell Growth Factors/pharmacology , Mice , Mice, Inbred DBA , Mice, Mutant Strains , Mice, SCID , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins c-kit/physiology , Receptors, Growth Factor/analysis , Receptors, Growth Factor/genetics , Recombinant Proteins/pharmacology , Sarcoma Viruses, Murine/genetics , Spleen/cytology , Spleen Focus-Forming Viruses/genetics , Stem Cell Factor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...