Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Oncogene ; 34(32): 4248-59, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-25381817

ABSTRACT

Obscurins, encoded by the single OBSCN gene, are giant cytoskeletal proteins with structural and regulatory roles. The OBSCN gene is highly mutated in different types of cancers. Loss of giant obscurins from breast epithelial cells confers them with a survival and growth advantage, following exposure to DNA-damaging agents. Here we demonstrate that the expression levels and subcellular distribution of giant obscurins are altered in human breast cancer biopsies compared with matched normal samples. Stable clones of non-tumorigenic MCF10A cells lacking giant obscurins fail to form adhesion junctions, undergo epithelial-to-mesenchymal transition and generate >100-µm mammospheres bearing markers of cancer-initiating cells. Obscurin-knockdown MCF10A cells display markedly increased motility as a sheet in 2-dimensional (2D) substrata and individually in confined spaces and invasion in 3D matrices. In line with these observations, actin filaments redistribute to extending filopodia where they exhibit increased dynamics. MCF10A cells that stably express the K-Ras oncogene and obscurin short hairpin RNA (shRNA), but not scramble control shRNA, exhibit increased primary tumor formation and lung colonization after subcutaneous and tail vein injections, respectively. Collectively, our findings reveal that loss of giant obscurins from breast epithelium results in disruption of the cell-cell contacts and acquisition of a mesenchymal phenotype that leads to enhanced tumorigenesis, migration and invasiveness in vitro and in vivo.


Subject(s)
Breast/metabolism , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Biopsy , Blotting, Western , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/genetics , Cell Line , Cell Movement/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Epithelium/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Intercellular Junctions/metabolism , Microscopy, Confocal , Neoplasm Metastasis , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rho Guanine Nucleotide Exchange Factors/metabolism , Transplantation, Heterologous , Vimentin/genetics , Vimentin/metabolism , ras Proteins/genetics , ras Proteins/metabolism
2.
Oncogene ; 34(35): 4558-69, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-25435370

ABSTRACT

Interstitial fluid flow in and around the tumor tissue is a physiologically relevant mechanical signal that regulates intracellular signaling pathways throughout the tumor. Yet, the effects of interstitial flow and associated fluid shear stress on the tumor cell function have been largely overlooked. Using in vitro bioengineering models in conjunction with molecular cell biology tools, we found that fluid shear (2 dyn/cm(2)) markedly upregulates matrix metalloproteinase 12 (MMP-12) expression and its activity in human chondrosarcoma cells. MMP-12 expression is induced in human chondrocytes during malignant transformation. However, the signaling pathway regulating MMP-12 expression and its potential role in human chondrosarcoma cell invasion and metastasis have yet to be delineated. We discovered that fluid shear stress induces the synthesis of insulin growth factor-2 (IGF-2) and vascular endothelial growth factor (VEGF) B and D, which in turn transactivate MMP-12 via PI3-K, p38 and JNK signaling pathways. IGF-2-, VEGF-B- or VEGF-D-stimulated chondrosarcoma cells display markedly higher migratory and invasive potentials in vitro, which are blocked by inhibiting MMP-12, PI3-K, p38 or JNK activity. Moreover, recombinant human MMP-12 or MMP-12 overexpression can potentiate chondrosarcoma cell invasion in vitro and the lung colonization in vivo. By reconstructing and delineating the signaling pathway regulating MMP-12 activation, potential therapeutic strategies that interfere with chondrosarcoma cell invasion may be identified.


Subject(s)
Bone Neoplasms/enzymology , Chondrosarcoma/enzymology , Insulin-Like Growth Factor II/physiology , Lung Neoplasms/enzymology , Matrix Metalloproteinase 12/metabolism , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor D/metabolism , Animals , Biomechanical Phenomena , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Chondrosarcoma/secondary , Enzyme Activation , Enzyme Induction , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/secondary , MAP Kinase Signaling System , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation
3.
Oncogene ; 32(14): 1761-71, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-22614020

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP1) interacts genetically with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) to suppress early-onset T-lineage lymphomas in the mouse, but the underlying mechanisms have remained unknown. To address this question, we analyzed a series of lymphomas arising in PARP1(-/-)/DNA-PKcs(-/-) (P1(-/-)/D(-/-)) mice. We found that, despite defective V(D)J recombination, P1(-/-)/D(-/-) lymphomas lacked clonal reciprocal translocations involving antigen-receptor loci. Instead, tumor cells were characterized by aneuploidy driven by two main mechanisms: p53 inactivation and abnormal chromosome disjunction due to telomere fusions (TFs). Aberrant accumulation of p53 was observed in 13/19 (68.4%) lymphomas. Sequence analysis revealed five p53 mutations: three missense point mutations (one transition in exon 8 and two transversions in exons 5 and 8, respectively), one in-frame 5-11 microindel in exon 7 and a 410-bp deletion encompassing exons 5-8, resulting in a truncated protein. Analysis of tumor metaphases using sequential telomere fluorescent in-situ hybridization and spectral karyotyping revealed that nine out of nine lymphomas contained TFs. Mutant but not wild-type p53 status was associated with frequent clonal and nonclonal TFs, suggesting that p53 normally limits the extent of telomere dysfunction during transformation. Chromosomes involved in TFs were more likely to be aneuploid than chromosomes not involved in TFs in the same metaphases, regardless of the p53 status, indicating that TFs promote aneuploidy via a mechanism that is distinct from p53 loss. Finally, analysis of radiation responses in P1(-/-)/D(-/-), and control primary cells and tissues indicates that loss of PARP1 increases in vivo radiosensitivity and genomic instability in DNA-PKcs-deficient mice without impairing p53 stabilization and effector functions, suggesting a more severe defect in double-strand break (DSB) repair in double mutants. Together, our findings uncover defective DSB repair leading to tumor suppressor inactivation and abnormal segregation of fused chromosomes as two novel mechanisms promoting tumorigenesis in thymocytes lacking PARP1 and DNA-PKcs.


Subject(s)
Cell Transformation, Neoplastic/pathology , DNA-Activated Protein Kinase/physiology , DNA-Binding Proteins/physiology , Lymphoma/etiology , Mutation/genetics , Nuclear Proteins/physiology , Poly(ADP-ribose) Polymerases/physiology , Telomere/physiology , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cesium Radioisotopes , Chromosome Aberrations , DNA/genetics , Female , Genomic Instability , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Lymphoma/metabolism , Lymphoma/pathology , Male , Mice , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Radiation Tolerance/genetics , Real-Time Polymerase Chain Reaction , Thymocytes/metabolism , Thymocytes/pathology , Translocation, Genetic , Tumor Cells, Cultured
4.
Oncogene ; 30(23): 2659-69, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21297660

ABSTRACT

Adenomatous polyposis coli (APC) gene mutations have been implicated in familial and sporadic gastrointestinal (GI) cancers. APC mutations are associated with autosomal dominant inheritance of disease in humans. Similarly, mice that contain a single mutant APC gene encoding a protein truncated at residue 716 (Apc(Δ716)) develop multiple polyps throughout the GI tract as early as 4 weeks after birth. Inactivation of another tumor suppressor gene, Hypermethylated in Cancer 1 (HIC1), often occurs in human colon cancers, among others, via CpG island hypermethylation. Homozygous deletion of Hic1 in mice results in major developmental defects and embryonic lethality. Hic1 heterozygotes have previously been shown to develop tumors of a variety of tissue types. We now report that loss of a single Hic1 allele can promote crypt hyperplasia and neoplasia of the GI tract, and Hic1(+/-), Apc(+/Δ716) double heterozygotes (DH) develop increased numbers of polyps throughout the GI tract at 60 days. Hic1 expression is absent in polyps from DH mice, with concomitant increased expression of two transcriptional repression targets of Hic1, Sirt1 and Sox9. Together, our data suggest that loss of a gene frequently silenced via epigenetic mechanisms, Hic1, can cooperate with loss of a gene mutated in GI cancer, Apc, to promote tumorigenesis in an in vivo model of multiple intestinal neoplasia.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/genetics , Intestine, Small/metabolism , Kruppel-Like Transcription Factors/genetics , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cells, Cultured , CpG Islands/genetics , DNA Methylation , Embryo, Mammalian/cytology , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Heterozygote , Humans , Hyperplasia , Immunohistochemistry , Intestine, Small/pathology , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
5.
Cytotherapy ; 7(6): 509-19, 2005.
Article in English | MEDLINE | ID: mdl-16306013

ABSTRACT

BACKGROUND: Human mesenchymal stem cells (hMSC) have been isolated and characterized extensively for a variety of clinical applications. Yet it is unclear how the phenomenon of hMSC plasticity can be safely and reasonably exploited for therapeutic use. METHODS: We have generated mesenchymal stem cells (MSC) from normal human BM and identified a novel cell population with a transformed phenotype. This cell population was characterized by morphologic, immunophenotypic, cytogenetic analyzes and telomerase expression. Its tumorigenicity in NOD/SCID mice was also studied. RESULTS: A subpopulation of cells in hMSC culture was noted to appear morphologically distinct from typical MSC. The cells were spherical, cuboidal to short spindle in shape, adherent and exhibited contact independent growth. Phenotypically the cells were CD133(+), CD34(-), CD45(-), CD90(low), CD105(-), VEGFR2(+). Cytogenetic analysis showed chromosome aneuploidy and translocations. These cells also showed a high level of telemerase activity compared with typical MSC. Upon transplantation into NOD/SCID mice, multiple macroscopic solid tumors formed in multiple organs or tissues. Histologically, these tumors were very poorly differentiated and showed aggressive growth with large areas of necrosis. DISCUSSION: The possible explanations for the origin of this cell population are: (1) the cells represent a transformed population of MSC that developed in culture; (2) abnormal cells existed in the donor BM at rare frequency and subsequently expanded in culture. In either case, the MSC culture may provide a suitable environment for transformed cells to expand or propagate in vitro. In summary, our data demonstrate the potential of transformed cells in hMSC culture and highlight the need for karyotyping as a release criteria for clinical use of MSC.


Subject(s)
Bone Marrow Cells/pathology , Mesenchymal Stem Cells/pathology , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , AC133 Antigen , Aneuploidy , Animals , Antigens, CD/analysis , Bone Marrow Cells/immunology , Cell Line, Transformed , Glycoproteins/analysis , Humans , Immunophenotyping , Karyotyping , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/enzymology , Neoplastic Stem Cells/immunology , Peptides/analysis , Time Factors
6.
Proc Natl Acad Sci U S A ; 98(26): 15155-60, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11724950

ABSTRACT

Current chemotherapeutic approaches for cancer are in part limited by the inability of drugs to destroy neoplastic cells within poorly vascularized compartments of tumors. We have here systematically assessed anaerobic bacteria for their capacity to grow expansively within avascular compartments of transplanted tumors. Among 26 different strains tested, one (Clostridium novyi) appeared particularly promising. We created a strain of C. novyi devoid of its lethal toxin (C. novyi-NT) and showed that intravenously injected C. novyi-NT spores germinated within the avascular regions of tumors in mice and destroyed surrounding viable tumor cells. When C. novyi-NT spores were administered together with conventional chemotherapeutic drugs, extensive hemorrhagic necrosis of tumors often developed within 24 h, resulting in significant and prolonged antitumor effects. This strategy, called combination bacteriolytic therapy (COBALT), has the potential to add a new dimension to the treatment of cancer.


Subject(s)
Bacteria, Anaerobic/growth & development , Neoplasms, Experimental/therapy , Animals , Antineoplastic Agents/therapeutic use , Clostridium/genetics , Clostridium/physiology , Combined Modality Therapy , Female , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasms, Experimental/drug therapy , Spores, Bacterial
7.
J Clin Invest ; 103(5): 691-6, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10074486

ABSTRACT

Chronic hypoxia induces polycythemia, pulmonary hypertension, right ventricular hypertrophy, and weight loss. Hypoxia-inducible factor 1 (HIF-1) activates transcription of genes encoding proteins that mediate adaptive responses to hypoxia, including erythropoietin, vascular endothelial growth factor, and glycolytic enzymes. Expression of the HIF-1alpha subunit increases exponentially as O2 concentration is decreased. Hif1a-/- mouse embryos with complete deficiency of HIF-1alpha due to homozygosity for a null allele at the Hif1a locus die at midgestation, with multiple cardiovascular malformations and mesenchymal cell death. Hif1a+/- heterozygotes develop normally and are indistinguishable from Hif1a+/+ wild-type littermates when maintained under normoxic conditions. In this study, the physiological responses of Hif1a+/- and Hif1a+/+ mice exposed to 10% O2 for one to six weeks were analyzed. Hif1a+/- mice demonstrated significantly delayed development of polycythemia, right ventricular hypertrophy, pulmonary hypertension, and pulmonary vascular remodeling and significantly greater weight loss compared with wild-type littermates. These results indicate that partial HIF-1alpha deficiency has significant effects on multiple systemic responses to chronic hypoxia.


Subject(s)
DNA-Binding Proteins/genetics , Hypoxia/genetics , Hypoxia/physiopathology , Nuclear Proteins/genetics , Transcription Factors , Animals , Blood Pressure , Heart Ventricles/physiopathology , Heterozygote , Homozygote , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mice
8.
Proc Natl Acad Sci U S A ; 96(2): 657-62, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9892689

ABSTRACT

NO. is a free radical that modulates heart function and metabolism. We report that a neuronal-type NO synthase (NOS) is located on cardiac sarcoplasmic reticulum (SR) membrane vesicles and that endogenous NO. produced by SR-associated NOS inhibits SR Ca2+ uptake. Ca2+-dependent biochemical conversion of L-arginine to L-citrulline was observed from isolated rabbit cardiac SR vesicles in the presence of NOS substrates and cofactors. Endogenous NO. was generated from the vesicles and detected by electron paramagnetic resonance spin-trapping measurements. Immunoelectron microscopy demonstrated labeling of cardiac SR vesicles by using anti-neuronal NOS (nNOS), but not anti-endothelial NOS (eNOS) or anti-inducible NOS (iNOS) antibodies, whereas skeletal muscle SR vesicles had no nNOS immunoreactivity. The nNOS immunoreactivity also displayed a pattern consistent with SR localization in confocal micrographs of sections of human myocardium. Western blotting demonstrated that cardiac SR NOS is larger than brain NOS (160 vs. 155 kDa). No immunodetection was observed in cardiac SR vesicles from nNOS knockout mice or with an anti-nNOS mu antibody, suggesting the possibility of a new nNOS-type isoform. 45Ca uptake by cardiac SR vesicles, catalyzed by Ca2+-ATPase, was inhibited by NO. produced endogenously from cardiac SR NOS, and 7-nitroindazole, a selective nNOS inhibitor, completely prevented this inhibition. These results suggest that a cardiac muscle nNOS isoform is located on SR of cardiac myocytes, where it may respond to intracellular Ca2+ concentration and modulate SR Ca2+ ion active transport in the heart.


Subject(s)
Myocardium/enzymology , Nitric Oxide Synthase/metabolism , Sarcoplasmic Reticulum/enzymology , Animals , Calcium/pharmacokinetics , Calcium-Transporting ATPases/antagonists & inhibitors , Electron Spin Resonance Spectroscopy , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Microscopy, Confocal , Myocardium/cytology , Nitric Oxide/metabolism , Rabbits
9.
Arch Biochem Biophys ; 353(2): 312-21, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9606965

ABSTRACT

While oxygen free radicals are important mediators of brain injury, questions remain regarding which cell types and enzyme pathways trigger this radical generation. Microglial cells have been hypothesized to be an important source of radical generation; however, the magnitude, kinetics, and mechanism of this process are unknown. Oxygen radical generation by stimulated primary microglia was directly measured and characterized by electron paramagnetic resonance spin trapping. Microglia, when stimulated by phorbol ester or opsonified zymosan, gave rise to EPR spectra characteristic of superoxide. Experiments performed in the presence of superoxide dismutase, catalase, deferoxamine, and dimethyl sulfoxide excluded generation of hydroxyl radicals in significant amounts. Microglial superoxide generation was blocked by the NADPH oxidase inhibitor diphenylene iodonium in a manner similar to that seen in neutrophils, suggesting that a neutrophil like NADPH oxidase was the source of superoxide production. However, microglia produced 20 to 40 times less superoxide compared to a similar number of neutrophils during the first 30 min following stimulation, indicating a marked difference in the regulation of NADPH oxidase activation. Western blots of microglia lysates demonstrated that both large (gp91-phox) and small (p22-phox) NADPH oxidase subunits are expressed in both unstimulated and stimulated microglia. Indirect immunofluorescence demonstrated localization at the membrane surfaces of activated cells. Thus, microglial cells generate superoxide via a neutrophil-like NADPH oxidase but exhibit distinctly different time course and magnitude of activation than that seen in neutrophils.


Subject(s)
Microglia/metabolism , NADPH Oxidases/metabolism , Superoxides/metabolism , Animals , Cattle , Cells, Cultured , Cyclic N-Oxides/metabolism , Electron Spin Resonance Spectroscopy , Fluorescent Antibody Technique, Indirect , Free Radicals , Sheep
10.
Viral Immunol ; 10(1): 15-20, 1997.
Article in English | MEDLINE | ID: mdl-9095528

ABSTRACT

A recombinant envelope glycoprotein derived from visna virus, a natural lentivirus pathogen of sheep, induced antibodies that neutralized cell-free virus and blocked virus-mediated cell-to-cell fusion. The visna virus envelope gene was subcloned into a baculovirus expression vector and was expressed in insect cells. A pair of guinea pigs were immunized with the recombinant glycoprotein, and postimmunization sera neutralized cell-free visna virus infectivity and also blocked virus-mediated syncytium formation when infected and uninfected cells were cocultured together. Thus, the recombinant visna surface envelope glycoprotein is capable of inducing both neutralizing and fusion-blocking antibodies. In addition to its relevance for vaccine development, the recombinant glycoprotein will be a useful tool to study differences between antibody-mediated enhancement versus neutralization during virus-host interactions in lentivirus infections in vivo.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Viral Envelope Proteins/immunology , Visna-maedi virus/immunology , Animals , Antigens, Viral/genetics , Cell Fusion , Cell Line , Cells, Cultured , Glycoproteins/genetics , Glycoproteins/immunology , Guinea Pigs , Macrophages/cytology , Macrophages/immunology , Neutralization Tests , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombination, Genetic , Spodoptera/cytology , Viral Envelope Proteins/genetics
11.
Vet Pathol ; 33(6): 692-6, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8952028

ABSTRACT

Two Hampshire-Duroc cross piglets maintained on 100% total parenteral nutrition (TPN) for 3 weeks developed pancreatic epithelial cell necrosis, diffuse acinar atrophy, and marked interstitial fibrosis. In addition, the piglets had severe villus atrophy in the small intestine as a result of TPN. Atomic absorption spectrophotometric analysis of liver samples revealed toxic hepatic zinc levels (513.5 and 491.2 ppm) in the TPN piglets (40-90 ppm in control piglets). Administering TPN bypasses homeostatic control mechanisms regulating zinc absorption at the gastrointestinal level and may reduce pancreatic secretion contributing to the accumulation of zinc in tissues. Intestinal villus atrophy, a sequela to TPN, may have also affected zinc excretion by impairing intestinal flux and desquamation. These factors should be considered in formulating TPN solutions and zinc levels administered must be reduced accordingly to avoid toxicity. Furthermore, sources and tissue levels of zinc should be investigated when necrosis, acinar atrophy, and fibrosis of the pancreas are encountered in young pigs.


Subject(s)
Pancreas/pathology , Pancreatic Diseases/veterinary , Parenteral Nutrition, Total/veterinary , Swine Diseases/pathology , Zinc/toxicity , Animals , Atrophy/pathology , Atrophy/veterinary , Epithelium/drug effects , Epithelium/pathology , Female , Fibrosis/pathology , Fibrosis/veterinary , Intestines/pathology , Intestines/ultrastructure , Liver/chemistry , Liver/pathology , Male , Microvilli/ultrastructure , Necrosis , Pancreas/drug effects , Pancreatic Diseases/etiology , Pancreatic Diseases/pathology , Random Allocation , Swine , Swine Diseases/etiology , Zinc/analysis
12.
Vet Pathol ; 33(4): 439-41, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8817845

ABSTRACT

A 13-year-old spayed female Siamese cat was submitted for necropsy following unsuccessful treatment for obstructive jaundice. Histopathologic examination revealed an adenocarcinoma of the hepatopancreatic ampulla. The carcinoma obstructed the pancreatic and common bile ducts entering the ampulla, resulting in severe diffuse acinar degeneration, atrophy and fibrosis of the pancreas, and dilatation of the bile ducts, biliary fibrosis, and ductule proliferation in the liver. In humans, carcinoma of the ampulla of Vater, the hepatopancreatic ampulla, is considered an uncommon malignancy.


Subject(s)
Adenocarcinoma/pathology , Adenocarcinoma/veterinary , Ampulla of Vater/pathology , Common Bile Duct Neoplasms/pathology , Common Bile Duct Neoplasms/veterinary , Animals , Cats , Female
14.
J Virol ; 63(4): 1811-3, 1989 Apr.
Article in English | MEDLINE | ID: mdl-2538659

ABSTRACT

Antibodies to visna virus neutralized the virus in fibroblasts and macrophages but specifically enhanced the binding, penetration, and uncoating of the virus in the latter cells. F(ab')2 fragments of the immune antibody neutralized the virus in fibroblasts but did not enhance the early stages of the virus life cycle in macrophages. Furthermore, these fragments did not neutralize infectivity in macrophages but delayed the appearance of infectious virus in cells after the inoculation of preincubated virus-F(ab')2 complexes.


Subject(s)
Macrophages/immunology , Virus Replication , Visna-maedi virus/immunology , Animals , Antigen-Antibody Complex , Immunoglobulin Fc Fragments/immunology , In Vitro Techniques , Neutralization Tests , Receptors, Fc/immunology , Sheep , Visna-maedi virus/growth & development
15.
J Virol ; 62(6): 1974-80, 1988 Jun.
Article in English | MEDLINE | ID: mdl-2835502

ABSTRACT

The lentivirus caprine arthritis-encephalitis virus (CAEV) is a pathogen of goats. It is transmitted in milk and causes a persistent infection in goats, which often fail to produce neutralizing antibodies to the virus. Native CAEV particles are remarkably resistant to digestion with proteinase K and are neutralized extremely slowly by immune sera. Our studies showed that the virus particles are heavily sialylated. Studies with highly specific sialyltransferase enzymes identified penultimate carbohydrate linkages typical of O- and N-linked oligosaccharides on the virus and suggested that the virus may be more heavily sialylated on O-linked than on N-linked oligosaccharides. Removal of sialic acids from the virus by neuraminidase treatment did not reduce infectivity of the particles. However, desialylation rendered the virus more susceptible to proteolysis by proteinase K. Desialylation also enhanced the kinetics of neutralization of the virus by goat antibodies. These results suggest that the carbohydrates on the viral surface are important both in protecting viral proteins from digestion by proteases and in protecting the virus from rapid neutralization by antibodies.


Subject(s)
Retroviridae/physiology , Sialic Acids/physiology , Endopeptidase K , Hemagglutination Inhibition Tests , Murine hepatitis virus/metabolism , Neuraminidase/metabolism , Neutralization Tests , Retroviridae/immunology , Serine Endopeptidases/metabolism , Sialyltransferases/metabolism , Visna-maedi virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...