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










Publication year range
1.
Vet Immunol Immunopathol ; 111(1-2): 81-95, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16549215

ABSTRACT

Equine herpesvirus-1 (EHV-1) is the cause of serious disease with high economic impact on the horse industry, as outbreaks of EHV-1 disease occur every year despite the frequent use of vaccines. Cytotoxic T-lymphocytes (CTLs) are important for protection from primary and reactivating latent EHV-1 infection. DNA vaccination is a powerful technique for stimulating CTLs, and the aim of this study was to assess antibody and cellular immune responses and protection resulting from DNA vaccination of ponies with combinations of EHV-1 genes. Fifteen ponies were divided into three groups of five ponies each. Two vaccination groups were DNA vaccinated on four different occasions with combinations of plasmids encoding the gB, gC, and gD glycoproteins or plasmids encoding the immediate early (IE) and early proteins (UL5) of EHV-1, using the PowderJect XR research device. Total dose of DNA/plasmid/vaccination were 25 microg. A third group comprised unvaccinated control ponies. All ponies were challenge infected with EHV-1 6 weeks after the last vaccination, and protection from clinical disease, viral shedding, and viremia was determined. Virus neutralizing antibodies and isotype specific antibody responses against whole EHV-1 did not increase in either vaccination group in response to vaccination. However, glycoprotein gene vaccinated ponies showed gD and gC specific antibody responses. Vaccination did not affect EHV-1 specific lymphoproliferative or CTL responses. Following challenge infection with EHV-1, ponies in all three groups showed clinical signs of disease. EHV-1 specific CTLs, proliferative responses, and antibody responses increased significantly in all three groups following challenge infection. In summary, particle-mediated EHV-1 DNA vaccination induced limited immune responses and protection. Future vaccination strategies must focus on generating stronger CTL responses.


Subject(s)
Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/immunology , Herpesvirus Vaccines/immunology , Horse Diseases/immunology , Horse Diseases/virology , Vaccines, DNA/immunology , Animals , Antibodies, Viral/blood , Cell Proliferation , Female , Genes, Immediate-Early/genetics , Genes, Immediate-Early/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Herpesvirus Vaccines/therapeutic use , Horse Diseases/prevention & control , Horses , Immunoglobulin Idiotypes/immunology , Male , Neutralization Tests/veterinary , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Vaccination/methods , Vaccination/veterinary , Vaccines, DNA/therapeutic use , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Virus Latency/immunology
2.
Vet Immunol Immunopathol ; 94(1-2): 47-62, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12842611

ABSTRACT

We have previously demonstrated that hemagglutinin (HA) gene vaccination and influenza virus infection generate protective antibody responses in equids. However, these antibody responses differ substantially in that particle mediated DNA vaccination does not induce an immunoglobulin A (IgA) response. A study was performed to investigate the regional immunoregulatory mechanisms associated with these different immune responses. Ponies were either vaccinated with equine HA DNA vaccines at skin and mucosal sites, infected with influenza virus or left untreated and influenza-specific antibody responses and protection from challenge infection was studied. In a subset of ponies, lymphocytes from peripheral blood (PBLs), nasopharyngeal mucosal tissue, or lymph nodes (LNLs) were collected for measurement of influenza virus-specific lymphoproliferative responses, local antibody production and IL-2, IL-4 and IFN-gamma mRNA production by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). DNA vaccination and influenza virus infection induced humoral immunoglobulin Ga (IgGa) and immunoglobulin Gb (IgGb) production and lymphoproliferative responses that were positively correlated with IFN-gamma mRNA production. However, there were marked differences in immune response in that only influenza infection induced an IgA response, and the regional distribution of lymphoproliferation, IFN-gamma and antibody responses. Responses to DNA vaccination occurred in PBLs and in lymph nodes draining DNA vaccination sites, while influenza virus infection induced responses in PBLs and hilar LNLs. In summary, common features of immune responses to either influenza virus infection or DNA vaccination were virus-specific IgGa, IgGb and IFN-gamma responses, which are associated with protection from infection, even when the regional distribution of these immune responses varied depending on the site of immune encounter.


Subject(s)
Antibodies, Viral/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/veterinary , Vaccines, DNA/immunology , Animals , Antibody Specificity , Cytokines/genetics , Cytokines/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Horse Diseases/immunology , Horse Diseases/virology , Horses/immunology , Horses/virology , Lymph Nodes/cytology , Lymph Nodes/immunology , Male , Orthomyxoviridae Infections/prevention & control , Virus Shedding
3.
Vaccine ; 21(21-22): 3081-92, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12798652

ABSTRACT

We have previously demonstrated that equine influenza virus hemagglutinin (HA) DNA vaccination protects ponies from challenge infection, and induces protective IgGa and IgGb responses. However, this approach does not induce a nasal IgA response. The objective of this study was to examine the value of cholera toxin (CT) administration as an adjuvant for intranasal HA DNA vaccination, and to measure protection 3 months after DNA vaccination. After an immunogenic dose of CT was determined, ponies were immunized on two occasions by intranasal administration of HA DNA and cholera toxin, or HA DNA alone. Ponies in both groups received two additional HA DNA particle mediated vaccinations at skin and mucosal sites. Antibody responses, and protection from challenge infection 3 months after the last vaccination were studied and compared to an influenza virus naive control group. Ponies in both vaccination groups produced virus-specific IgG antibodies in serum following vaccination and showed clinical protection from challenge infection 3 months after the last vaccination. Co-administration of CT plus HA DNA vaccination induced a nasal IgA response. In addition, analysis of antibody titers in nasal secretions indicated local production of nasal IgGb, which was amplified by CT administration.


Subject(s)
Cholera Toxin/immunology , Hemagglutinins, Viral/genetics , Immunity, Mucosal , Immunoglobulin A/biosynthesis , Influenza Vaccines/immunology , Vaccines, DNA/immunology , Administration, Intranasal , Animals , Antibodies, Viral/blood , Cholera Toxin/administration & dosage , Female , Horse Diseases/immunology , Horse Diseases/prevention & control , Horse Diseases/virology , Horses , Influenza A virus , Influenza Vaccines/administration & dosage , Male , Nasal Mucosa/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/veterinary , Vaccination/veterinary , Vaccines, DNA/administration & dosage
4.
Clin Cancer Res ; 7(4): 952-61, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309346

ABSTRACT

Particle-mediated gene delivery was used to immunize mice against melanoma. Mice were immunized with a plasmid cDNA coding for the human melanoma-associated antigen, gp100. Murine B16 melanoma, stably transfected with human gp100 expression plasmid, was used as a tumor model. Particle-mediated delivery of gp100 plasmid into the skin of naïve mice resulted in significant protection from a subsequent tumor challenge. Co-delivery of murine granulocyte-macrophage colony-stimulating factor (GM-CSF) expression plasmid together with the gp100 plasmid consistently resulted in a greater level of protection from tumor challenge. The inclusion of the GM-CSF plasmid with the gp100 DNA vaccine allowed a reduction in the gp100 plasmid dose required for antitumor efficacy. Protection from tumor challenge was achieved with as little as 62.5 ng of gp100 DNA per vaccination. Tumor protection induced by the gp100 + GM-CSF gene combination was T cell mediated, because it was abrogated in vaccinated mice treated with anti-CD4 and anti-CD8 monoclonal antibodies. In addition, administration of the gp100 + GM-CSF DNA vaccine to mice bearing established 7-day tumors resulted in significant suppression of tumor growth. These results indicate that inclusion of GM-CSF DNA augments the efficacy of particle-mediated vaccination with gp100 DNA, and this form of combined gp100 + GM-CSF DNA vaccine warrants clinical evaluation in melanoma patients.


Subject(s)
DNA/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Melanoma, Experimental/prevention & control , Membrane Glycoproteins/therapeutic use , Neoplasm Proteins/therapeutic use , Animals , DNA/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Melanoma, Experimental/drug therapy , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Neoplasm Proteins/genetics , Plasmids/genetics , Recurrence , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Vaccination , gp100 Melanoma Antigen
5.
Scand J Plast Reconstr Surg Hand Surg ; 34(4): 301-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11195866

ABSTRACT

We have used an in vitro model of wound contraction, the fibroblast-populated collagen lattice, to examine the effect of platelet-derived growth factor BB (PDGF-BB) and PDGF-BB gene transfer by gene gun on the contraction of lattices composed of either diabetic or non-diabetic human fibroblasts. The area of collagen lattice and DNA synthesis were measured in 12 specimens. There were significant increases in lattice contraction with increasing doses of PDGF-BB and fibroblasts transfected with the PDGF-BB gene compared with control (p < 0.01). DNA synthesis of the non-diabetic and diabetic fibroblast lattices showed significantly increased incorporation of tritiated thymidine with increasing doses of PDGF-BB and fibroblasts transfected with the PDGF-BB compared with controls (p < 0.05). The effect of PDGF-BB gene transfer on diabetic and non-diabetic fibroblasts was similar to that of 20 ng/ml or less of PDGF-BB.


Subject(s)
Biolistics , Collagen/ultrastructure , Diabetes Mellitus, Type 2/pathology , Fibroblasts/physiology , Platelet-Derived Growth Factor/genetics , Skin/cytology , Wound Healing/physiology , Becaplermin , Cells, Cultured , DNA/biosynthesis , Humans , Proto-Oncogene Proteins c-sis , Transfection
6.
Methods Mol Med ; 29: 297-303, 2000.
Article in English | MEDLINE | ID: mdl-21374329

ABSTRACT

Particle-mediated delivery involves coating materials onto the surface of dense sub-cellular sized (0.5-5 mm) particles and accelerating the particles to sufficient velocity to penetrate target cells. The technique was invented by Sanford and Wolf at Cornell University (1) to transfer DNA into intact plant cells (2), and was further developed into an effective process for producing genetically engineered crop plants by several groups (reviewed in 3). Subsequent work has shown that this method is generally applicable for transferring materials including DNA, RNA, proteins, peptides and pharmacological compounds into a wide variety of tissue and cell types in vivo, ex vivo, or in vitro (reviewed in 4).

7.
Vaccine ; 17(18): 2245-58, 1999 May 04.
Article in English | MEDLINE | ID: mdl-10403592

ABSTRACT

Equine influenza virus infection remains one of the most important infectious diseases of the horse, yet current vaccines offer only limited protection. The equine immune response to natural influenza virus infection results in long-term protective immunity, and is characterized by mucosal IgA and serum IgGa and IgGb antibody responses. DNA vaccination offers a radical alternative to conventional vaccines, with the potential to generate the same protective immune responses seen following viral infection. Antigen-specific antibody isotype responses in serum and mucosal secretions were studied in ponies following particle-mediated delivery of hemagglutinin (HA)-DNA vaccination on three occasions at approximately 63-day intervals. One group of four ponies were vaccinated at skin and mucosal sites and the another group were vaccinated at skin sites only. All ponies were subjected to a challenge infection 30 days after the third vaccination. Skin and mucosal vaccination provided complete protection from clinical signs of infection, while skin vaccination provided partial protection; DNA vaccination provided partial protection from viral shedding. DNA vaccination generated only IgGa and IgGb antibody responses, which occurred with a higher frequency in the skin and mucosa vaccinated ponies. No mucosal IgA response was generated prior to challenge infection and IgA responses were only detected in those ponies which shed virus postchallenge. These results demonstrate that HA-DNA vaccination induces IgG(a) and IgG(b) antibody responses which are associated with protection in the absence of mucosal IgA responses. In addition, additional DNA vaccinations of mucosal sites increased protection and the frequency of seroconversion in ponies.


Subject(s)
Antibodies, Viral/biosynthesis , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Horse Diseases/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/veterinary , Vaccines, DNA/immunology , Animals , Female , Horse Diseases/virology , Horses , Influenza A virus/chemistry , Influenza A virus/immunology , Influenza A virus/isolation & purification , Influenza Vaccines/genetics , Male , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control
8.
J Surg Res ; 78(2): 85-91, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9733623

ABSTRACT

BACKGROUND: Gene transfer to skin has many potential applications but lacks a safe, practical delivery method. This report presents a new technique, microseeding, for in vivo gene transfer to skin and wounds and for DNA-mediated vaccination. The plasmid DNA solution was delivered directly to the target cells of the skin by a set of oscillating solid microneedles driven by a modified tattooing device. MATERIALS AND METHODS: Skin and partial-thickness excisional wounds in pigs were microseeded with either hEGF expression plasmid or beta-galactosidase expression plasmid. Human EGF was also delivered by single injection or particle bombardment. hEGF expression in wound fluid and in target tissue was determined by ELISA with anti-hEGF-specific antibodies. Additionally, weanling pigs were microseeded with a hemagglutinin of swine influenza virus expression plasmid and production of anti-HA-specific antibodies was determined by blocking ELISA. RESULTS: hEGF expression in microseeded partial thickness wounds (5664 pg/site) and skin sites (969 pg/site) peaked 2 days after transfection being four- to seven-fold higher than gene transfer by a single intradermal injection and two- to three-fold higher than particle-mediated gene transfer. The beta-galactosidase-expressing cells were detected in dermis and epidermis. Pigs microseeded with HA expression plasmid were protected from infection by the Swine influenza virus. CONCLUSIONS: These results demonstrate that microseeding is a simple and effective method for in vivo gene transfer to skin and wounds and is more efficient than single injection and particle-mediated gene transfer.


Subject(s)
Gene Transfer Techniques , Influenza A virus , Orthomyxoviridae Infections , Plasmids , Wound Healing/genetics , Animals , Epidermal Growth Factor/genetics , Female , Gene Expression Regulation, Enzymologic , Histocompatibility Antigens/genetics , Skin Physiological Phenomena , Swine , Tattooing/methods , Vaccination/methods , beta-Galactosidase/genetics
9.
J Virol ; 72(2): 1491-6, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9445052

ABSTRACT

Particle-mediated delivery of a DNA expression vector encoding the hemagglutinin (HA) of an H1N1 influenza virus (A/Swine/Indiana/1726/88) to porcine epidermis elicits a humoral immune response and accelerates the clearance of virus in pigs following a homotypic challenge. Mucosal administration of the HA expression plasmid elicits an immune response that is qualitatively different than that elicited by the epidermal vaccination in terms of inhibition of the initial virus infection. In contrast, delivery of a plasmid encoding an influenza virus nucleoprotein from A/PR/8/34 (H1N1) to the epidermis elicits a strong humoral response but no detectable protection in terms of nasal virus shed. The efficacy of the HA DNA vaccine was compared with that of a commercially available inactivated whole-virus vaccine as well as with the level of immunity afforded by previous infection. The HA DNA and inactivated viral vaccines elicited similar protection in that initial infection was not prevented, but subsequent amplification of the infection is limited, resulting in early clearance of the virus. Convalescent animals which recovered from exposure to virulent swine influenza virus were completely resistant to infection when challenged. The porcine influenza A virus system is a relevant preclinical model for humans in terms of both disease and gene transfer to the epidermis and thus provides a basis for advancing the development of DNA-based vaccines.


Subject(s)
DNA, Viral/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Viral Vaccines/immunology , Animals , Immunization , Orthomyxoviridae Infections/prevention & control , Swine
10.
Hybridoma ; 17(6): 569-76, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9890713

ABSTRACT

Class-switched, affinity-matured murine monoclonal antibody (MAb)-producing cell lines were generated against the Flt-3 receptor in less than 4 weeks following polynucleotide immunizations, used in conjunction with repetitive immunizations, multiple sites (RIMMS). Plasmid DNA encoding Flt-3/Fc was coated onto gold particles, which were subsequently propelled into the epidermis of mice using biolistic particle bombardment using the Accell gene gun. Pools of immune peripheral lymph node cells were somatically fused 13 days after the onset of delivery of DNA encoding the target antigen. To determine if early responses could be augmented, DNA-encoding murine GM-CSF was delivered 3 days prior to the Flt-3/Fc DNA immunizations. The data presented demonstrates the successful identification and characterization of class-switched, affinity-matured MAbs that bind to the Flt-3 receptor. When compared to conventional methodologies or intramuscular targeted DNA-based immunization for the generation of MAbs, use of the gene gun in conjunction with RIMMS allows for a more rapid production of affinity-matured MAb-producing cell lines.


Subject(s)
Antibodies, Monoclonal/immunology , Immunization/methods , Membrane Proteins/immunology , Animals , Antibody Formation , Biolistics , DNA/genetics , DNA/immunology , Humans , Membrane Proteins/genetics , Mice
11.
Hybridoma ; 16(4): 381-9, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9309429

ABSTRACT

Affinity matured murine monoclonal antibody producing cell lines can now be rapidly generated using a novel repetitive, multiple site immunization strategy designated RIMMS. RIMMS capitalizes on rapid hypermutation and affinity maturation events which occur in B cell populations localized within secondary lymphatic tissue early in response to antigenic challenges. A murine myeloma cell line, P3XBcl-2-13, stably transfected with Bcl-2, enhances the outgrowth of hybridomas following somatic fusion with immune lymphocytes isolated from pooled peripheral lymph nodes (PLN) 8-14 days after the initial immunization. Immunizations somatic fusion, screening and isolation of affinity matured IgG secreting monoclonal antibody cell lines occur within a one month time period. By using RIMMS, we have been able to expedite the isolation of affinity matured monoclonal antibodies to numerous antigens, including a drug hapten.


Subject(s)
Antibodies, Monoclonal/immunology , B-Lymphocytes/immunology , Hybridomas/immunology , Animals , Antibody Affinity , Antibody Specificity , Blotting, Western , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Genes, bcl-2/genetics , Haptens/immunology , Humans , Immunization , Mice , Precipitin Tests , Tumor Cells, Cultured
12.
Vaccine ; 15(10): 1149-56, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9269061

ABSTRACT

Two fundamentally different approaches to vaccination of BALB/c mice with the hemagglutinin (HA) of A/Equine/Kentucky/1/81 (H3N8) (Eq/KY) were evaluated, that is, administration of HA protein vs administration of HA-encoding DNA. Each vaccine was tested for its immunogenicity and ability to provide protection from homologous virus challenge. HA protein was synthesized in vitro by infection of Sf21 insect cells with a recombinant baculovirus. Intranasal administration of this vaccine induced virus-specific antibodies, as measured by enzyme-linked immunosorbent assay (ELISA), but did not induce virus neutralizing (VN) antibodies. This route of administration provided partial protection from virus challenge, but interestingly, this protection was completely abrogated, rather than enhanced, by co-administration of 10 micrograms of cholera holotoxin. As a second approach, mice were directly vaccinated in vivo by Accell gene gun delivery of plasmid DNA encoding the Eq/KY HA gene. This approach induced VN antibodies as well as virus-specific ELISA antibodies. When two doses of DNA vaccine were administered 3 weeks apart, mice were not protected from challenge, although they cleared the infection more rapidly than control mice. However, when the second DNA vaccination was delayed until 9 weeks after the first, 9 out of 10 vaccinated mice were completely protected. These results indicate that the time between initial and booster DNA vaccinations may be an important variable in determining DNA vaccination efficacy.


Subject(s)
Influenza A virus/immunology , Influenza Vaccines/pharmacology , Vaccines, DNA/pharmacology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/blood , Baculoviridae/genetics , Base Sequence , Cholera Toxin/administration & dosage , DNA Primers/genetics , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Immunization Schedule , Immunization, Secondary , Influenza A virus/genetics , Influenza A virus/isolation & purification , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Lung/virology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Polymerase Chain Reaction , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
13.
Behring Inst Mitt ; (98): 73-8, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9382772

ABSTRACT

Polynucleotide vaccines are a new approach to immunization that promises qualitative advances in vaccine technology. These vaccines mimic infection in that they result in expression of pathogen gene products in situ, which can elicit both cell-mediated immune responses and humoral responses. This approach has been applied primarily to vaccines against viral diseases, but may be significant for vaccines directed toward bacterial pathogens. Auragen has developed a generally applicable gene transfer technology and, for vaccine applications, has focused on particle-mediated gene transfer to epidermis. Results demonstrate that Accell polynucleotide vaccines induce immune responses toward human immunodefficiency virus (HIV) antigens, influenza A virus antigens, and hepatitis B virus (HBV) antigens in rodent,s swine and primates. Cellular immune responses toward these antigens have been demonstrated in rodents. In a swine influenza a challenge model Accell vaccination provides protection equivalent to that of a commercial killed-whole-virus vaccine. Vaccination of mice by this method toward a Chlamydia pneumoniae major outer-membrane protein elicits a species-specific antibody response.


Subject(s)
Bacterial Vaccines , Gene Transfer Techniques , Vaccines, DNA , Viral Vaccines , AIDS Vaccines , Animals , Antibody Formation , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Hepatitis B Vaccines , Humans , Immunity, Mucosal , Influenza Vaccines , Mice , Primates , Skin/immunology , Swine
14.
J Infect Dis ; 175(1): 7-15, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8985190

ABSTRACT

Two people developed symptoms of influenza 36 h after collecting nasal swabs from pigs experimentally infected with A/Sw/IN/1726/88 (Sw/IN). Pharyngeal swabs from these persons tested positive for influenza virus RNA 8 days after infection. Analysis of hemi-nested polymerase chain reaction (PCR) products indicated that the hemagglutinin (HA) segments of the isolates were genetically related to the HA of Sw/IN. Four influenza A virus isolates (A/WI/4754/94, A/WI/4756/94, A/WI/4758/94, A/WI/4760/94) were recovered from a 39-year-old man and 2 (A/WI/4755/94, A/WI/4757/94) from a 31-year-old woman. The HAs of the isolates were antigenically indistinguishable from the virus used to infect the pigs. Sequence analysis of the HA genes indicated they were 99.7% identical to the HA of the virus used in the experiment. Multisegment reverse transcription-PCR proved that all of the segments originated from Sw/IN, demonstrating that transmission of swine H1N1 viruses to humans occurs directly and readily, despite Animal Biosafety Level 3 containment practices used for these experiments.


Subject(s)
Influenza A virus/isolation & purification , Influenza, Human/transmission , Influenza, Human/virology , Swine Diseases/virology , Adult , Animals , Cloning, Molecular , Female , Genome, Viral , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A virus/classification , Influenza A virus/genetics , Influenza A virus/immunology , Influenza, Human/veterinary , Male , Molecular Sequence Data , Pharynx/virology , Polymerase Chain Reaction , Sequence Analysis , Swine , Zoonoses
15.
Proc Natl Acad Sci U S A ; 91(25): 12188-92, 1994 Dec 06.
Article in English | MEDLINE | ID: mdl-7991604

ABSTRACT

This report details the transfer of a human epidermal growth factor (hEGF) expression plasmid to porcine partial-thickness wound keratinocytes by particle-mediated DNA transfer (Accell). After gene transfer an external sealed fluid-filled wound chamber was used to protect the wound, provide containment of the exogenous DNA and expressed peptide, and permit sampling of the wound fluid. Analysis of wound fluid for hEGF and total protein, an indicator of reformation of the epithelial barrier, showed that wounds bombarded with the hEGF plasmid exhibited a 190-fold increase in EGF concentration and healed 20% (2.1 days) earlier than the controls. EGF concentrations in wound fluid persisted over the entire 10-day monitored period, decreasing from 200 pg/ml to 25 pg/ml over the first 5 days. Polymerase chain reaction results showed that plasmid DNA was present in the wound for at least 30 days. These findings demonstrate the possible utility of in vivo gene transfer to enhance epidermal repair.


Subject(s)
Epidermal Growth Factor/biosynthesis , Gene Transfer Techniques , Keratinocytes/physiology , Wound Healing , Wounds and Injuries/physiopathology , Amino Acid Sequence , Animals , Base Sequence , Biological Assay , Cell Division , DNA/analysis , DNA Primers , Epidermal Growth Factor/analysis , Epidermal Growth Factor/physiology , Exons , Humans , KB Cells , Molecular Sequence Data , Plasmids , Polymerase Chain Reaction , Recombinant Fusion Proteins/biosynthesis , Swine , Time Factors , Wounds and Injuries/therapy
16.
EMBO J ; 12(4): 1621-31, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8467809

ABSTRACT

Theileria parva is an obligate, intracellular, parasitic protozoan that causes East Coast fever, an acute leukemia-like disease of cattle. T. parva and the related parasite, Theileria annulata, are unique among protozoa in that their intralymphocytic stages induce transformation of bovid lymphocytes. Comparison of in vitro protein kinase activities between uninfected IL-2-dependent T lymphoblasts and T. parva-infected lymphocytes revealed a 4.7- to 12-fold increase in total phosphorylation and the induction of a group of Theileria infection-specific phosphoproteins. The enzyme that phosphorylates these substrates is a serine/threonine kinase with substrate and effector specificities of casein kinase (CK) II. Northern blot analyses revealed a 3.9- to 6.0-fold increase in CKII alpha mRNA in the infected cells relative to the controls. Furthermore, a marked increase of CKII antigen was observed on Western blots of materials prepared from the infected cell lines. The antibovine CKII antibody used in these studies immunoprecipitated a protein kinase that phosphorylated casein in a reaction that was inhibited by low (nM) quantities of heparin. Our data show marked increases of bovine CKII at the transcriptional, translational and functional levels in T. parva-infected lymphocytes, relative to quiescent cells or IL-2-dependent parental lymphoblasts. Bovine CKII thus appears to be constitutively activated in these cells and we propose that this kinase may be an important element in the signal-transducing pathways activated by Theileria in bovid lymphocytes and perhaps in some leukemic cells.


Subject(s)
Lymphocytes/parasitology , Protein Serine-Threonine Kinases/biosynthesis , Theileria parva/pathogenicity , Theileriasis/enzymology , Animals , Base Sequence , Casein Kinase II , Cattle , Enzyme Induction , Gene Expression , Heparin/pharmacology , Lymphocytes/enzymology , Molecular Sequence Data , Phosphoproteins/metabolism , Polyamines/pharmacology , Precipitin Tests , RNA, Messenger/genetics
17.
Biochemistry ; 31(27): 6193-202, 1992 Jul 14.
Article in English | MEDLINE | ID: mdl-1378299

ABSTRACT

Theileria parva is an obligate intracellular protozoan parasite which is the causative agent of East Coast fever, an acute, leukemia-like disease of cattle. The intralymphocytic stage of the parasite induces blastogenesis and clonal expansion of quiescent bovid lymphocytes. Experiments in our laboratory have shown a marked increase of casein kinase II- (CK II-) like activity in T. parva-transformed lymphocytes. We have also detected CK II activity in purified T. parva schizonts. To explore the significance of this increase, we used a Drosophila melanogaster CK II alpha cDNA probe [Saxena et al. (1987) Mol. Cell Biol. 7, 3409-3417] to isolate a T. parva genomic clone encoding a CK II catalytic subunit. The clone contains a 1.3-kb open reading frame coding for a predicted protein of 420 amino acids (M(r) 50,200). Northern blot analysis revealed a single transcript of 1.65 kb. The deduced T. parva CK II catalytic subunit sequence shows, over 321 residues comprising the C-terminus of the molecule, extensive identity with CK II alpha and alpha' sequences from both vertebrate and invertebrate organisms. The T. parva CK II subunit amino acid sequence displays 68% identity with the Drosophila alpha subunit and 67% with the Caenorhabditis elegans alpha subunit but only 58% and 56% sequence identity with the Saccharomyces cerevisiae alpha and alpha' subunits, respectively. Comparison of the T. parva sequence with higher eukaryotic alpha and alpha' sequences reveals that it is most identical with the alpha subunit. A unique component of the T. parva CK II alpha subunit is a 99 amino acid sequence at the N-terminus, which contains a sequence motif with features characteristic of signal peptides.


Subject(s)
Lymphocytes/immunology , Protein Serine-Threonine Kinases/genetics , Theileria parva/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Blotting, Southern , Casein Kinase II , Cattle , Cloning, Molecular , DNA/genetics , DNA/isolation & purification , Genes , Genomic Library , Lymphocyte Activation , Lymphocytes/parasitology , Macromolecular Substances , Molecular Sequence Data , RNA/genetics , RNA/isolation & purification , Restriction Mapping , Sequence Homology, Nucleic Acid , Theileria parva/isolation & purification , Theileria parva/pathogenicity
18.
Mol Cell Biol ; 4(12): 2643-52, 1984 Dec.
Article in English | MEDLINE | ID: mdl-6084810

ABSTRACT

Ribosomal (r) proteins encoded by polyadenylated RNA were specifically precipitated in vitro from polysomes by using antibodies raised against characterized Drosophila melanogaster r proteins. The immuno-purified mRNA in the polysome complex was used to prepare cDNA with which to probe a D. melanogaster genomic library. Selected recombinant phages were used to hybrid select mRNAs, which were analyzed by in vitro translation. Three clones containing the genes for r proteins 7/8, S18, and L12 were positively identified by electrophoresis of the translation products in one-dimensional and two-dimensional polyacrylamide gels. Sequences encoding r proteins S18 and L12 were found to be present in the genome in single copies. In contrast, the polynucleotide containing the region encoding 7/8 may be repeated or may contain or be flanked by short repeated sequences. The sizes of mRNAs that hybridized to the recombinant clone containing 7/8 were significantly larger than would be expected from the molecular weight of protein 7/8, implying that there were unusually long 5' and 3' noncoding sequences. The mRNAs for r proteins S18 and L12 were however, only about 10% larger. In situ hybridizations to salivary gland polytene chromosomes, using the recombinant phage, revealed that the recombinant clone containing the gene for r protein 7/8 hybridized to 5D on the X chromosome; the recombinant clone containing the gene for S18 hybridized to 15B on the same chromosome, and the recombinant phage containing the gene for L12 hybridized to 62E on chromosome 3L. It is of interest that the genomic locations of all three r protein clones were within the chromosomal intervals known to contain the Minute mutations [M(1)0, M(1)30, and M(3)LS2]. Although each clone contained sequences specifying two to four proteins, none had more than one identifiable r protein gene, suggesting that different D. melanogaster r protein genes may not be closely linked.


Subject(s)
Cloning, Molecular , DNA/isolation & purification , Drosophila melanogaster/genetics , Ribosomal Proteins/genetics , Animals , Base Sequence , Chromosome Mapping , DNA Restriction Enzymes/metabolism , Electrophoresis, Polyacrylamide Gel , Nucleic Acid Hybridization , Poly A/metabolism , RNA/metabolism , RNA, Messenger
19.
Eur J Biochem ; 127(1): 199-205, 1982 Sep.
Article in English | MEDLINE | ID: mdl-6814910

ABSTRACT

The relatively acidic proteins (group A80) of Drosophila melanogaster ribosomes were separated by ion-exchange chromatography. Fractions containing one or more acidic proteins were combined into thirteen pools. The criterion for the combination was the migration pattern in one-dimensional polyacrylamide gels containing sodium dodecyl sulphate. Five proteins (7/8, S25/S27, S14, L1/L2 and L5/L6) required no further purification. The others were further purified as follows: proteins S7, and S9 by preparative gel electrophoresis: and protein 13 (to newly identified protein) by adsorption with conconavalin-A--agarose. Four proteins had no detectable contamination, and in each of the others the impurities were no greater than 3%. The amount of purified protein recovered from a starting amount of 2.63 g total 80-S ribosomal protein and a starting amount of 105 mg group A80 varied from 0.4 mg to 8.8 mg. The molecular weight of the proteins was estimated by polyacrylamide gel electrophoresis in sodium dodecyl sulphate. The amino acid composition of the individual purified proteins was determined. Several phosphorylated proteins were identified. Proteins 13b and 13c are phosphorylated derivatives of 13a; 7b/8b and 7c/8c are phosphorylated derivatives of 7a and/or 8a. Proteins 7/8 and 13 are distinct proteins but are very similar in amino acid composition.


Subject(s)
Drosophila melanogaster/analysis , Ribosomal Proteins/isolation & purification , Amino Acids/analysis , Animals , Chemical Phenomena , Chemistry , Hydrogen-Ion Concentration , Phosphorylation
20.
Mol Gen Genet ; 187(3): 370-4, 1982.
Article in English | MEDLINE | ID: mdl-6817033

ABSTRACT

Antibodies raised against D. melanogaster ribosomal proteins were used to examine possible structural relationships between eukaryotic and prokaryotic ribosomal proteins. The antisera were raised against either groups of ribosomal proteins or purified individual ribosomal proteins from D. melanogaster. The specificity of each antiserum was confirmed and the identity of the homologous E. coli ribosomal protein was determined by immunochemical methods. Immuno-overlay assays indicated that the antiserum against the D. melanogaster small subunit protein S14 (anti-S14) was highly specific for protein S14. In addition, anti-S14 showed a cross-reaction with total E. coli ribosomal proteins in Ouchterlony double immunodiffusion assays and with only E. coli protein S6 in immuno-overlay assays. From these and other experiments with adsorption of anti-S14 with individual purified proteins, the E. coli protein homologous to the D. melanogaster protein S14 was established as protein S6.


Subject(s)
Drosophila melanogaster/genetics , Escherichia coli/genetics , Ribosomal Proteins/genetics , Animals , Electrophoresis, Polyacrylamide Gel , Immunodiffusion , Ribosomal Proteins/isolation & purification , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...