Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Int J Clin Exp Pathol ; 7(3): 858-69, 2014.
Article in English | MEDLINE | ID: mdl-24696707

ABSTRACT

Coxsackievirus B (CVB) is a significant pathogen that causes pediatric central nervous system disease with acute syndromes commonly. The onset of its infection was abrupt, and after recovery there usually will be severe mental sequelae. The disease model for research was not established by the way of natural infection, although there are various investigations about the CVB-induced central nervous system (CNS) diseases. Thus, we have established an acute neonatal CNS disease mice model by CVB orally infecting. This model imitated the natural infection route and focuses the onset of CNS disease, inducing severe infection and lesion in the hippocampus and cortex regions, and the stability of the model was demonstrated. A pathology score system was developed for quantitative pathology analysis, which standardizes the CNS pathology analysis by statistics analysis. By this model, the track of CVB penetrating the blood brain barrier in vivo has been captured. One of the experimental strains CVB3/Macocy, as a new variant, was isolated, and its genomic RNA was cloned. According to its nucleotide sequence, we have characterized its genomic structure and defined its genotype. Based on the sequence, some mutations which do not change the CVB-induced CNS damage have been found. The model is an effective tool for studies on CVB-induced CNS diseases.


Subject(s)
Central Nervous System Diseases/virology , Coxsackievirus Infections/pathology , Disease Models, Animal , Enterovirus/genetics , Animals , Animals, Newborn , Brain/pathology , Brain/virology , Central Nervous System Diseases/pathology , DNA, Viral/genetics , DNA, Viral/isolation & purification , Female , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C
2.
Int J Clin Exp Pathol ; 7(3): 890-904, 2014.
Article in English | MEDLINE | ID: mdl-24696708

ABSTRACT

Coxsackievirus B (CVB) is a significant pathogen of neonatal diseases with severe systemic involvement and high mortality. Hence, it is essential to develop a CVB-induced acute systemic disease model on newborn mouse and study the injury at the onset phase. In this work, a clinical strain of CVB3, Nancy, and its variant strain, Macocy, were adopted in 24 hour old neonates by oral infection. The pathological changes in the heart, liver and lung tissues were analyzed by pathology assays. In situ end labeling assay for programmed cell death was carried out for liver tissues. The data on fatality and infection rates and pathology scores were analyzed statistically. The genomic sequences of the two strains were aligned. The model represented the manifest clinical syndromes of hepatitis, pneumonia and myocardial injury at the onset phase, in which massive numbers of hepatocytes had undergone programmed cell death. Statistical and pathological analysis indicated that the myocardial injury was mild, whereas the liver and lung were more severe. The fatality rate, infection and pathology of the two CVB strains were the same. Therefore, two nucleotide mutations in the 5' UTR and four amino acid mutations in polyprotein, which did not alter virulence, were shown. By peroral CVB infection of neonatal mice, we developed an acute systemic disease model for studying visceral pathology and systemic disease. At the onset of acute neonatal systemic disease, the hepatitis and pneumonia may be the dominant reason of death, as the injury of liver and lung is more severe than that of heart.


Subject(s)
Coxsackievirus Infections/pathology , Enterovirus/pathogenicity , Liver Diseases/pathology , Lung Diseases/pathology , Animals , Animals, Newborn , Apoptosis/physiology , Coxsackievirus Infections/genetics , Disease Models, Animal , Enterovirus/genetics , Female , Heart Diseases/pathology , Heart Diseases/virology , Immunohistochemistry , In Situ Nick-End Labeling , Liver Diseases/virology , Lung Diseases/virology , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RNA, Viral/genetics
3.
Hybridoma (Larchmt) ; 31(4): 279-83, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22894782

ABSTRACT

The G10P[15] rotavirus CC0812-1 isolated from a diarrheal woman in Wuhan, China, in 2008 is phylogenetically close to the Lanzhou lamb rotavirus (LLR) of a monovalent human rotavirus vaccine produced by the Lanzhou Institute of Biological Products, China, and rotavirus Lamb-NT. This rotavirus can be used as the backbone of the attenuated rotavirus reassortant as a rotavirus vaccine candidate. In this study, rotavirus CC0812-1 was purified from the culture supernatant of CC0812-1-infected MA104 cells and used as antigen to immunize BALB/c mice. Four hybridoma clones were developed secreting antibodies that reacted with CC0812-1, designated as 1B1, 1B8, 1F11, and 1G10, respectively. Western blot analysis indicated that the four monoclonal antibodies (MAbs) were all specific for VP4 of rotavirus CC0812-1. Isotyping revealed that MAbs 1B1, 1B8, and 1G10 belonged to the IgM class, while MAb 1F11 belonged to the IgG1 subclass. A neutralization test demonstrated that the four MAbs all had the capacity to neutralize rotavirus CC0812-1. The neutralizing titers of the BALB/c mice ascites were 1:2048, 1:1024, 1:512, and 1:512 for MAbs 1B1, 1B8, 1F11, and 1G10, respectively.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/biosynthesis , Antibodies, Neutralizing/biosynthesis , Capsid Proteins/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neutralizing/immunology , Capsid/immunology , Capsid/ultrastructure , Cell Line , Female , Hybridomas , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Mice , Mice, Inbred BALB C , Rotavirus/immunology , Rotavirus/ultrastructure
4.
Virol J ; 7: 126, 2010 Jun 13.
Article in English | MEDLINE | ID: mdl-20540804

ABSTRACT

BACKGROUND: Bluetongue virus (BTV) is an icosahedral non-enveloped virus within the genus Orbivirus of Reoviridae and exists as 24 distinct serotypes. BTV can infect all ruminant species and causes severe sickness in sheep. Recently, it was reported that BTV can infect some human cancer cells selectively. Because of the important oncolysis of this virus, we developed a novel purifying method for large-scale production. The purifying logic is simple, which is picking out all the components unwanted and the left is what we want. The process can be summarized in 4 steps: centrifugation, pulling down cell debrises and soluble proteins by co-immunoprecipitation with agarose Protein A, dialysis and filtration sterilization after concentration. RESULTS: The result of transmission electron microscope (TEM) observation showed that the sample of purified virus has a very clear background and the virions still kept intact. The result of 50% tissue culture infective dose (TCID(50)) assay showed that the bioactivity of purified virus is relatively high. CONCLUSIONS: This method can purify BTV-10 with high quality and high biological activity on large-scale production. It also can be used for purifying other BTV serotypes.


Subject(s)
Bluetongue virus/isolation & purification , Bluetongue/virology , Immunoprecipitation/methods , Staphylococcal Protein A/chemistry , Virology/methods , Animals , Bluetongue virus/physiology , Bluetongue virus/ultrastructure , Chlorocebus aethiops , Protein Binding , Sepharose/chemistry , Vero Cells
5.
Microbiol Immunol ; 54(12): 754-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21223364

ABSTRACT

Whether administration of total alkaloids from Commelina communis L. (TAC) reduces lung damage in influenza virus-infected mice was investigated. Compared with untreated mice, significantly less severe damage was found in the lungs of mice administered TAC at 8 mg/kg per day for 6 days. TAC significantly decreased viral loads in the lungs. The concentrations of IFN-γ in the serum of TAC-treated mice were significantly lower than those of virus control mice at 4 and 6 days post-infection. The results indicate that TAC imparted partial protection to the mice by reducing pulmonary viral loads and limiting lesions in the lungs.


Subject(s)
Alkaloids/therapeutic use , Commelina/chemistry , Lung/pathology , Orthomyxoviridae Infections/drug therapy , Animals , Female , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Phytotherapy , Tumor Necrosis Factor-alpha/biosynthesis
6.
Arch Virol ; 154(11): 1837-40, 2009.
Article in English | MEDLINE | ID: mdl-19774337

ABSTRACT

The antiviral activity of total alkaloids from Commelina communis L. (TAC) against influenza virus A/PR/8/34 (H1N1) was investigated in vitro and in vivo. TAC exhibited an inhibitory action on the growth of influenza virus in Madin-Darby canine kidney cells when added before or after viral infection. In mice infected with influenza virus, orally administered TAC at 8, 16 or 32 mg/kg per day for 6 days significantly increased the survival rate, prolonged the mean survival time and reduced the viral titers in the lung and the lung index, compared with that of the untreated virus control. The results obtained suggest that TAC has a pronounced protective effect against infection by influenza A virus.


Subject(s)
Alkaloids/chemistry , Alkaloids/pharmacology , Commelina/chemistry , Influenza A Virus, H1N1 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Dogs , Female , Mice , Mice, Inbred BALB C , Specific Pathogen-Free Organisms
7.
Acta Oncol ; 47(1): 124-34, 2008.
Article in English | MEDLINE | ID: mdl-17851866

ABSTRACT

Bluetongue viruses (BTVs) infect primarily domestic cattle and wild ruminants but have never been shown to infect normal human cells. Thus, humans are sero-negative towards BTVs. The selective and differential effects of BTV serotype 10 (BTV-10) infection were investigated with five cell lines including primary human embryo lung fibroblast (HEL) and primary murine embryos fibroblast(MEF), human hepatic carcinoma 3B cell line (Hep-3B), human lung carcinoma cell line (A549) and mouse fibroblast cell line (NIH 3T3). In this study, comparative analyses of differential cytopathic effects (CPEs), survival rates using different Multiplicities of Infection (MOI), ultra-structural changes by transmission electron microscopy, and the preferential cell cycle changes of infected cells by flow cytometry were made among these cells. Detection of the presence of BTV genome and kinetic analysis of virus titers in TCID50 were also made. We provided the first analytical demonstration and evidence that BTV-10 could selectively infect and degrade human cancer cells but not cultured primary normal cells. No CPE or viral mRNAs could be detected within these normal cells, while various degrees of CPE could be found in Hep-3B and A549, as well as in NIH 3T3 under similar conditions. Before death, BTV-infected human cancer cells were directly arrested in the sub-G1 phase and the diversity of BTV infection as shown by the MTT method had significant difference (F = 95.635, p < 0.01). Above results suggested that this viral dose-dependent cytotoxic effect is caused by both effective virion amplification and induced apoptosis. Cellular distinctive transformation status may contribute to the selectivity. Thus, selective degradation of human cancer cells but not normal diploid cells by the newly discovered oncolytic potential of BTV would provide a very attractive approach for cancer therapy in the future.


Subject(s)
Apoptosis , Bluetongue virus , G1 Phase , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Carcinoma/virology , Carcinoma, Hepatocellular/virology , Cattle , Cell Line , Cell Line, Tumor , Fibroblasts/virology , Flow Cytometry , Humans , In Vitro Techniques , Liver Neoplasms/virology , Lung Neoplasms/virology , Mice , Microscopy, Electron, Transmission , NIH 3T3 Cells
8.
Zhonghua Zhong Liu Za Zhi ; 29(7): 505-9, 2007 Jul.
Article in Chinese | MEDLINE | ID: mdl-18069629

ABSTRACT

OBJECTIVE: To study the death mode of human hepatocellular carcinoma Hep-3B cells and human lung adenocarcinoma A549 cells induced by bluetongue virus strain HbC3 (BTV-HbC3) and the mechanism of its action. METHODS: BTV-HbC3 was used to infect the tumor cells, and the cytopathic effects (CPE) was observed. TUNEL staining was used to detect the apoptosis of tumor cells induced by BTV-HbC3. The changes of endoplasmic reticulum and nuclei treated with BTV-HbC3 were further examined by laser scanning confocal microscopy. The activities of caspase-3/7, caspase-8 and caspase-9 were determined by fluorescence analysis. RESULTS: Hep-3B cells were sensitive to BTV-HbC3. Lots of early apoptotic cells were found by TUNEL staining. The laser scanning confocal microscopic examination showed characteristics of apoptosis, such as pyknotic nuclei, margination of nuclear chromatin and vacuolization of endoplasmic reticulumin in Hep-3B cells exposed to BTV-HbC3. The activity of caspase-3/7 was increased, but the activity changes of caspase-8 and caspase-9 were not found. A549 cells were sensitive to BTV-HbC3 too. But no apoptotic cells were observed by TUNEL staining. The results of laser scanning confocal microscopy showed marked vacuolization of endoplasmic reticulum, but chromatin margination was not found after A549 cells was exposed to BTV-HbC3. The activity of caspase-3/7 and caspase-9 was increased, but the activity of caspase-8 was not changed. CONCLUSION: BTV-HbC3 induces apoptosis of Hep-3B tumor cells mainly through endoplasmic reticulum signal transduction pathway, and the features of cell death in A549 cells could be described as paraptosis.


Subject(s)
Apoptosis , Bluetongue virus/pathogenicity , Liver Neoplasms/virology , Lung Neoplasms/virology , Oncolytic Viruses/pathogenicity , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/virology , Bluetongue virus/physiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Nucleus/pathology , Endoplasmic Reticulum/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oncolytic Viruses/physiology , Signal Transduction
9.
Acta Pharmacol Sin ; 28(4): 493-502, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17376288

ABSTRACT

AIM: Angiopoietin-1 (Ang1) and vascular endothelial growth factor A (VEGF) play important roles in vascular formation and maturation, suggesting a combination of these 2 would be a promising therapy for ischemic diseases. So we constructed an adeno-associated virus (AAV) vector, simultaneously encoding human VEGF(165) and Ang1 (AAV-VEGF/Ang1), and investigated its therapeutic effect in a rabbit ischemic hind-limb model. METHODS: Four experimental groups were used to prepare the rabbit ischemic hind-limb model following AAV vectors intramuscular administration as follows: PBS (phosphate buffered solution), AAV-VEGF, AAV-Ang1, AAV-VEGF/Ang1. RESULTS: Eight weeks after administration, human VEGF(165) and Ang1 were detected by RT-PCR, Western blotting and histochemical staining methods in AAV-VEGF/Ang1 transduced muscles. Group AAV-VEGF/Ang1 showed a significantly increased blood-flow recovery in ischemic hind-limbs compared with the other groups. Histological staining for alkaline phosphatase showed that capillary density of group AAV-VEGF/Ang1 or AAV-VEGF was significantly higher than that of group PBS or AAV-Ang1. Histological immunostaining for smooth muscle alpha-actin (alpha-SMA) revealed that group AAV-VEGF/Ang1 had the highest density of alpha-SMA-positive vessels compared with the other groups. Vascular leakage, one of the major adverse effects induced by VEGF, was very severe in group AAV-VEGF, but the permeability was obviously reduced when VEGF was co-expressed with Ang1 in group AAV-VEGF/Ang1. CONCLUSION: AAV vectors can simultaneously encode 2 proteins which can be efficiently and stably co-expressed in transduced tissues. AAV-mediated VEGF and Ang1 gene transfer enhances neovascularization, prevents capillary leakage, and improves blood flow in a rabbit hind-limb ischemic model. These findings suggest that intramuscular administration of AAV-VEGF/Ang1 may be useful in the treatment of ischemic diseases.


Subject(s)
Angiopoietin-1/biosynthesis , Angiopoietin-1/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Hindlimb/blood supply , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Animals , Capillary Permeability/genetics , Capillary Permeability/physiology , Hindlimb/physiology , Humans , Ischemia/pathology , Male , Plasmids/genetics , Rabbits , Regional Blood Flow/genetics , Regional Blood Flow/physiology
10.
Cancer Biol Ther ; 4(11): 1234-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16357508

ABSTRACT

The selectively oncolytic effects of mtHSV, a HSV icp34.5 mutant with lacz gene insertion, on several tumor cells in vitro and its antitumor effects by the intratumoral (IT) route to nude mice loaded the human hepatoma xenografts were explored. The mtHSV could conditionally replicate in and lyse Hep-3B (human hepatoma cells), Hep-2 (human larynx cancer cells) and SPC-A1 (human lung cancer cells), but not MRC-5 (human fibroblast cells). The 125 nude mice loaded with Hep-3B were randomly divided into five treatment groups and given three IT injections with three different dose of the mtHSV, adriamycin (ADM), or vehicle (supernatant of non-infected Vero cells). Significant tumor growth inhibition (30%-70%) was seen in the nude mice treated IT with mtHSV, whereas tumors treated IT with Vero supernatant displayed rapid tumor growth. The results of regular and biochemical blood examination, systemic necropsy and pathological slices showed that mtHSV almost has no side effect on treated mice. RT-PCR results revealed that the replication of mtHSV was exclusively confined to the treated tumors, but not to other organs. Our results provide further preclinical evidence that mtHSV may be used as an oncolytic agent for cancer therapy.


Subject(s)
Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Neoplasms/therapy , Neoplasms/virology , Xenograft Model Antitumor Assays , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Cells, Cultured , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Liver Neoplasms/virology , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Transplantation, Heterologous , Virus Replication
11.
Intervirology ; 48(5): 292-6, 2005.
Article in English | MEDLINE | ID: mdl-15956796

ABSTRACT

We studied the sensitivity of several human cancer cell strains (HeLa, HT1080, SPC-A1, and ACHN) and normal cell strains (MRC-5 and Wish) to mumps virus (MuV) S79, a live attenuated vaccine strain. These cells exhibited a differential sensitivity to infection with MuV, and the susceptible sequences were ACHN > HeLa > HT1080 > SPC-A1 > Wish > MRC-5. In experiments in vivo, nude mice with HT1080 fibrosarcoma xenografts were randomly divided into three groups for intratumoral treatment with MuVS79, UV-inactivated MuVS79, and PBS. At 10(7) PFU, live MuVS79 injection caused in 7 of 9 mice complete regression by day 15 while rapid tumor growth occurred in all 9 mice treated with PBS. Rapid tumor progression also occurred in all 8 mice treated with UV-inactivated virus; however, tumor growth was delayed in the logarithmic phase relative to the PBS-treated tumors.


Subject(s)
Fibrosarcoma/therapy , Mumps virus/physiology , Neoplasms/therapy , Neoplasms/virology , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Fibrosarcoma/virology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/pathology , Virus Replication
12.
J Virol Methods ; 117(2): 129-36, 2004 May.
Article in English | MEDLINE | ID: mdl-15041209

ABSTRACT

Two obstacles limiting the efficacy of nearly all cancer gene therapy trails are low gene transduction efficiency and the lack of tumor specificity. Fortunately, a replication-competent, E1B-deficient adenovirus (dl1520) was developed that could overcome these limitations, because it was capable of efficiently and selectively destroying tumor cells lacking functional p53. In an attempt to appraise the efficiency and safety of this approach, a novel recombinant adenovirus, r3/Ad, containing a gfp-zeocin expression cassette was constructed in this work. The study in vitro demonstrated that r3/Ad has the ability to replicate in and lyse only the p53-deficient human tumor cells such as the human glioblastoma cells (U251) and human bladder cells (EJ) but not in the human fibroblast cells (MRC-5) with functional p53. Importantly, this gfp-zeocin fusion gene driven by the bipromoter (CMV and EM-7) could be used as an effective selective marker and reporter in prokaryotic and eukaryotic cells; and also zeocin as a selective marker could minimize contamination of the recombinant virus by the wt-Ad5. Additionally, it was found that the r3/Ad could be useful for studying the selective replication of E1B-deficient adenovirus in vivo, it could be used as a "guide" to study the ability of the recombinant adenovirus to spread and to infect distant tumor cells in any tumor bearing animal model by GFP as a reporter. This may help determine the safety of using any E1B-deficient adenovirus in cancer gene therapy.


Subject(s)
Adenoviridae/genetics , Tumor Suppressor Protein p53/deficiency , Bleomycin , Cell Line, Tumor , Cloning, Molecular , Cytomegalovirus/genetics , Genes, p53 , Genetic Vectors , Green Fluorescent Proteins , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Microscopy, Fluorescence , Mutagenesis, Insertional , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/genetics , Recombination, Genetic , Virus Replication
13.
J Biochem Mol Biol ; 36(4): 379-86, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12895296

ABSTRACT

A mutant herpes simplex virus 1, mtHSV, was constructed by inserting the E. coli beta-galactosidase gene into the loci of icp34.5, the apoptosis-inhibiting gene of HSV. The mtHSV replicated in and lysed U251 (human glioma cells), EJ (human bladder cells), and S-180 (mice sarcoma cells), but not Wish (human amnion cells) cells. With its intact tk (thymidine kinase) gene, mtHSV exhibited susceptibility to acyclovir (ACV), which provided an approach to control viral replication. An in vivo test with mtHSV was conducted in immune-competent mice bearing sarcoma S-180 tumors, which were treated with a single intratumoral injection of mtHSV or PBS. Tumor dimensions then were measured at serial time points, and the tumor volumes were calculated. Sarcoma growth was significantly inhibited with prolonged time and reduced tumor volume. There was microscopic evidence of necrosis of tumors in treated mice, whereas no damage was found in other organs. Immunohistochemical staining revealed that virus replication was exclusively confined to the treated tumor cells. HSV-1 DNA was detected in tumors, but not in the other organs by a polymerase chain reaction analysis. From these experiments, we concluded that mtHSV should be a safe and promising oncolytic agent for cancer treatment.


Subject(s)
Genetic Therapy , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Sarcoma 180/therapy , Viral Proteins/genetics , Acyclovir/pharmacology , Animals , Antiviral Agents/pharmacology , Apoptosis , Cell Line , Cell Line, Tumor , Gene Transfer Techniques , Herpesvirus 1, Human/drug effects , Humans , Mice , Mice, Inbred BALB C , Mutation , Necrosis , Neoplasm Transplantation , Sarcoma 180/pathology , Sarcoma 180/virology , Thymidine Kinase/metabolism , Virus Replication/drug effects
14.
J Biochem Mol Biol ; 36(3): 275-81, 2003 May 31.
Article in English | MEDLINE | ID: mdl-12787482

ABSTRACT

An E1B-defective adenovirus, named r2/Ad carrying the neo expression cassette, was constructed by homologous recombination. The construction, selection (using neomycin as a selective marker), and propagation of the recombinant virus was performed in human embryonic kidney 293 cells (HEK 293). An in vitro study demonstrated that this recombinant virus has the ability to replicate in and lyse some p53-deficient human tumor cells such as human glioma tumor cells (U251) and human bladder cells (EJ), but not in some cells with functional p53, such as human adenocarcinoma cells (A549) and human fibroblast cells (MRC-5). Also, based on the cytopathic effect (CPE), it was demonstrated, under identical conditions, that the U251 cells were more sensitive to r2/Ad replication than the EJ cells. In this paper, we report that r2/Ad could be very useful in studying the in vitro selective replication of E1B-defective adenovirus and has great potential in cancer gene therapy.


Subject(s)
Adenoviridae/genetics , Kanamycin Kinase/genetics , Neomycin/pharmacology , Virus Replication/genetics , Adenoviridae/drug effects , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/metabolism , Cell Line , Cell Line, Tumor , Drug Resistance, Microbial/genetics , Electrophoresis, Polyacrylamide Gel , Genetic Markers , Genetic Vectors/genetics , Humans , Kanamycin Kinase/metabolism , Recombination, Genetic , Tumor Suppressor Protein p53/genetics , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...