Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Protein Expr Purif ; 70(2): 158-62, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19796689

ABSTRACT

Mass vaccination against anthrax with existing vaccines is costly and unsafe due to potential side effects. For post-infection treatment, passive immunotherapy measures are currently available, most based on anthrax protective antigen (PA)-specific therapeutic antibodies. Efficient against wild-type strains, these treatment(s) might fail to protect against infections caused by genetically engineered Bacillus anthracis strains. A recent discovery revealed that the von Willebrand factor A (VWA) domain of human capillary morphogenesis protein 2 (CMG2) is an exceptionally effective anthrax toxin receptor (ATR) proficient in helping to resolve this issue. Here we describe in planta production of chimeric recombinant protein (immunoadhesin) comprised of functional ATR domain fused with the human immunoglobulin Fc fragment (pATR-Fc). The fusion design allowed us to obtain pATR-Fc in plant green tissues in a soluble form making it fairly easy to purify by Protein-A chromatography. Standardized pATR-Fc preparations (purity>90%) were shown to efficiently bind anthrax PA as demonstrated by ELISA and Western blot analysis. Recombinant pATR-Fc was also shown to protect J774A1 macrophage cells against the anthrax toxin. This study confirmed that plant-derived pATR-Fc antibody-like protein is a prospective candidate for anthrax immunotherapy.


Subject(s)
Immunoglobulin Fc Fragments/genetics , Membrane Proteins/genetics , Receptors, Peptide/genetics , Recombinant Fusion Proteins/biosynthesis , Bacillus anthracis/immunology , Humans , Membrane Proteins/biosynthesis , Plants, Genetically Modified/genetics , Receptors, Peptide/biosynthesis , Nicotiana/genetics
2.
J Hum Hypertens ; 24(5): 359-62, 2010 May.
Article in English | MEDLINE | ID: mdl-19865105

ABSTRACT

The role of uric acid (UA) in human physiology is subject to controversy. Either it is an important radical scavenger, a mostly neutral, waste metabolic product that may cause gout and kidney stones if elevated, or it is involved in the causation of hypertension, vascular and renal diseases. Recently we conducted a clinical trial to determine whether raising the serum UA levels through the oral administration of inosine is well tolerated and may benefit patients with multiple sclerosis. An important aspect of the safety profile is whether raising the serum UA levels elevates blood pressure. During the 1-year trial, blood pressure and serum UA levels were monitored in 16 patients. Both parameters were recorded throughout the trial that included 69 visits by patients at baseline and during the placebo phase as well as 138 visits while receiving inosine treatment. We have observed that although the serum UA levels increased significantly during the inosine treatment phase of the trial, from 4.2+/-0.8 to 7.1+/-1.7 mg per 100 ml, blood pressure remained unchanged, averaging 123+/-15/78+/-9. Our findings indicate that raising the serum UA levels to upper normal physiological levels for a period of up to 1-year does not influence blood pressure significantly.


Subject(s)
Blood Pressure/drug effects , Inosine/pharmacology , Multiple Sclerosis/blood , Multiple Sclerosis/physiopathology , Uric Acid/blood , Administration, Oral , Adult , Blood Pressure/physiology , Cross-Over Studies , Double-Blind Method , Female , Humans , Inosine/administration & dosage , Male , Middle Aged , Treatment Outcome
3.
Vaccine ; 27(28): 3730-4, 2009 Jun 08.
Article in English | MEDLINE | ID: mdl-19464556

ABSTRACT

The current diphtheria-tetanus-pertussis (DTP) pediatric vaccine is produced from the corresponding pathogenic bacteria Corynebacterium diphtheriae, Clostridium tetani and Bordetella pertussis; five injected doses of DTaP (acellular) vaccine are required for every child in the standard US vaccination schedule. Because the vaccine is derived from native live sources, adverse effects are possible and production is complex and costly. To address issues of safety, ease of renewability and expense, we used recombinant technology in an effort to develop a subunit DPT vaccine derived in non-pathogenic plant expression systems. Expression of diphtheria toxin (DT), tetanus fragment-C (TetC) and the non-toxic S1 subunit of pertussis toxin (PTX S1) antigenic proteins in soluble form in low-alkaloid tobacco plants and carrot cell cultures allowed efficient downstream purification to levels suitable for intramuscular injection in BALB/c mice. At working concentrations of 5mug per dose, these preparations induced high levels of antigen-specific IgGs in mouse sera. Our results clearly support the feasibility of producing recombinant pediatric vaccine components in plants.


Subject(s)
Diphtheria-Tetanus-Pertussis Vaccine/biosynthesis , Diphtheria-Tetanus-Pertussis Vaccine/immunology , Plants, Genetically Modified/metabolism , Animals , Antibodies, Bacterial/blood , Daucus carota/genetics , Daucus carota/metabolism , Diphtheria Toxin/biosynthesis , Diphtheria Toxin/genetics , Diphtheria Toxin/immunology , Diphtheria-Tetanus-Pertussis Vaccine/genetics , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Peptide Fragments/immunology , Pertussis Toxin/biosynthesis , Pertussis Toxin/genetics , Pertussis Toxin/immunology , Plants, Genetically Modified/genetics , Tetanus Toxin/biosynthesis , Tetanus Toxin/genetics , Tetanus Toxin/immunology , Nicotiana/genetics , Nicotiana/metabolism , United States , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/genetics , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics
4.
Vaccine ; 27(9): 1289-92, 2009 Feb 25.
Article in English | MEDLINE | ID: mdl-19162113

ABSTRACT

Polypeptide variants of the HA1 antigenic domain of the H5N1 avian influenza virus hemagglutinin (HA) molecule were produced in plants using transient and stable expression systems and fused with His/c-myc tags or with mouse or human Fc antibody fragments. The resulting peptides were purified and used for intramuscular immunization of mice. While the recombinant HA1 variants induced a significant serum humoral immune response in the mice, none of the HA1 preparations induced virus-neutralizing antibodies. Fusion with the Fc fragment improved overall yield of the constructs and allowed purification requiring only a single step, but led to no detectable fusion-related enhancement of immunogenicity or quality of immune response.


Subject(s)
Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza in Birds/immunology , Plant Proteins/immunology , Animals , Antibody Formation , Birds , Enzyme-Linked Immunosorbent Assay , Female , Humans , Influenza in Birds/genetics , Mice , Mice, Inbred BALB C
5.
Curr Top Microbiol Immunol ; 318: 325-42, 2008.
Article in English | MEDLINE | ID: mdl-18219824

ABSTRACT

In the past decade, a growing number of evidence has implicated free radicals in a variety of pathophysiological conditions including aging, cancer, and coronary heart disease. Analyses of different aspects of multiple sclerosis (MS) pathology with respect to oxidative damage have also revealed evidence of free radical injury to the central nervous system (CNS), although attempts to protect the CNS using various antioxidants have met with only moderate success. Several recent studies have reported lower levels of uric acid (UA), a major scavenger of reactive nitrogen species, in MS patients, while other studies found no such correlation. Here, we discuss these studies as well as current efforts to manipulate serum UA levels in MS patients.


Subject(s)
Inosine/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Uric Acid/metabolism , Blood-Brain Barrier , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Female , Free Radicals/metabolism , Humans , Inosine/administration & dosage , Male , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Treatment Outcome , Uric Acid/administration & dosage , Uric Acid/blood
6.
Vaccine ; 20(25-26): 3155-64, 2002 Aug 19.
Article in English | MEDLINE | ID: mdl-12163267

ABSTRACT

A new approach to the production and delivery of vaccine antigens is the use of engineered amino virus-based vectors. A chimeric peptide containing antigenic determinants from rabies virus glycoprotein (G protein) (amino acids 253-275) and nucleoprotein (N protein) (amino acids 404-418) was PCR-amplified and cloned as a translational fusion product with the alfalfa mosaic virus (AlMV) coat protein (CP). This recombinant CP was expressed in two plant virus-based expression systems. The first one utilized transgenic Nicotiana tabacum cv. Samsun NN plants providing replicative functions in trans for full-length infectious RNA3 of AlMV (NF1-g24). The second one utilized Nicotiana benthamiana and spinach (Spinacia oleracea) plants using autonomously replicating tobacco mosaic virus (TMV) lacking native CP (Av/A4-g24). Recombinant virus containing the chimeric rabies virus epitope was isolated from infected transgenic N. tabacum cv. Samsun NN plants and used for parenteral immunization of mice. Mice immunized with recombinant virus were protected against challenge infection. Based on the previously demonstrated efficacy of this plant virus-based experimental rabies vaccine when orally administered to mice in virus-infected unprocessed raw spinach leaves, we assessed its efficacy in human volunteers. Three of five volunteers who had previously been immunized against rabies virus with a conventional vaccine specifically responded against the peptide antigen after ingesting spinach leaves infected with the recombinant virus. When rabies virus non-immune individuals were fed the same material, 5/9 demonstrated significant antibody responses to either rabies virus or AlMV. Following a single dose of conventional rabies virus vaccine, three of these individuals showed detectable levels of rabies virus-neutralizing antibodies, whereas none of five controls revealed these antibodies. These findings provide clear indication of the potential of the plant virus-based expression systems as supplementary oral booster for rabies vaccinations.


Subject(s)
Glycoproteins/immunology , Nicotiana/metabolism , Nucleoproteins/immunology , Rabies Vaccines/biosynthesis , Rabies virus/immunology , Spinacia oleracea/metabolism , Viral Proteins/immunology , Administration, Oral , Alfalfa mosaic virus/genetics , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Capsid Proteins/physiology , Defective Viruses/genetics , Food , Glycoproteins/biosynthesis , Glycoproteins/genetics , Humans , Mice , Mice, Inbred C3H , Neutralization Tests , Nucleoproteins/biosynthesis , Nucleoproteins/genetics , Plant Leaves , Plants, Genetically Modified/metabolism , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies Vaccines/isolation & purification , Rabies virus/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Species Specificity , Spinacia oleracea/genetics , Nicotiana/genetics , Tobacco Mosaic Virus/genetics , Vaccination/methods , Vaccines, Subunit/biosynthesis , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification , Viral Proteins/biosynthesis , Viral Proteins/genetics
7.
Mult Scler ; 7(5): 313-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11724447

ABSTRACT

Peroxynitrite has been implicated in the pathogenesis of multiple sclerosis (MS) and its animal model experimental allergic encephalomyelitis (EAE). Previously, we have shown that administration of uric acid (UA), a peroxynitrite scavenger, is therapeutic in EAE We have also shown that MS patients have lower levels of serum uric acid than healthy individuals or those with other neurological diseases. The aim of this investigation was therefore to raise serum UA levels in MS patients. Oral administration of UA failed to increase low serum UA levels, evidently due to its degradation by gastrointestinal bacteria. However, serum UA could be raised and maintained at elevated levels for a year and more without reported side-effects by oral administration of its precursor inosine. Three of 11 patients given inosine showed some evidence of clinical improvement and there was no sign of disease progression in the remaining patients. Gadolinium-enhanced lesions, observed in two patients before receiving inosine, could not be detected after either 10 or IS months inosine treatment These data provide evidence that serum UA levels can be readily manipulated and that the benefit of higher levels to individuals with MS should be studied further in greater number of patients.


Subject(s)
Inosine/administration & dosage , Multiple Sclerosis, Chronic Progressive/drug therapy , Multiple Sclerosis, Chronic Progressive/metabolism , Peroxynitrous Acid/metabolism , Administration, Oral , Adult , Female , Gadolinium , Humans , Inosine/pharmacokinetics , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis, Chronic Progressive/pathology , Peroxynitrous Acid/antagonists & inhibitors , Uric Acid/blood , Uric Acid/cerebrospinal fluid
8.
J Virol ; 75(22): 10800-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602721

ABSTRACT

The pathogenicity of individual rabies virus strains appears to correlate inversely with the extent of apoptotic cell death they induce and with the expression of rabies virus glycoprotein, a major inducer of an antiviral immune response. To determine whether the induction of apoptosis by rabies virus contributes to a decreased pathogenicity by stimulating antiviral immunity, we have analyzed these parameters in tissue cultures and in mice infected with a recombinant rabies virus construct that expresses the proapoptotic protein cytochrome c. The extent of apoptosis was strongly increased in primary neuron cultures infected with the recombinant virus carrying the active cytochrome c gene [SPBN-Cyto c(+)], compared with cells infected with the recombinant virus containing the inactive cytochrome c gene [SPBN-Cyto c(-)]. Mortality in mice infected intranasally with SPBN-Cyto c(+) was substantially lower than in SPBN-Cyto c(-)-infected mice. Furthermore, virus-neutralizing antibody (VNA) titers were significantly higher in mice immunized with SPBN-Cyto c(+) at the same dose. The VNA titers induced by these recombinant viruses paralleled their protective activities against a lethal rabies virus challenge infection, with SPBN-Cyto c(+) revealing an effective dose 20 times lower than that of SPBN-Cyto c(-). The strong increase in immunogenicity, coupled with the marked reduction in pathogenicity, identifies the SPBN-Cyto c(+) construct as a candidate for a live rabies virus vaccine.


Subject(s)
Cytochrome c Group/biosynthesis , Rabies virus/enzymology , Animals , Antibodies, Viral/blood , Female , Immunization , Mice , Mice, Inbred C3H , Nucleocapsid/analysis , Nucleocapsid Proteins , Phenotype , Rabies Vaccines/immunology , Rabies virus/genetics , Rabies virus/immunology , Recombination, Genetic
9.
J Immunol ; 167(6): 3470-7, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11544340

ABSTRACT

We have recently demonstrated that increased blood-CNS barrier permeability and CNS inflammation in a conventional mouse model of experimental allergic encephalomyelitis are dependent upon the production of peroxynitrite (ONOO(-)), a product of the free radicals NO* and superoxide (O2*(-)). To determine whether this is a reflection of the physiological contribution of ONOO(-) to an immune response against a neurotropic pathogen, we have assessed the effects on adult rats acutely infected with Borna disease virus (BDV) of administration of uric acid (UA), an inhibitor of select chemical reactions associated with ONOO(-). The pathogenesis of acute Borna disease in immunocompetent adult rats results from the immune response to the neurotropic BDV, rather than the direct effects of BDV infection of neurons. An important stage in the BDV-specific neuroimmune response is the invasion of inflammatory cells into the CNS. UA treatment inhibited the onset of clinical disease, and prevented the elevated blood-brain barrier permeability as well as CNS inflammation seen in control-treated BDV-infected rats. The replication and spread of BDV in the CNS were unchanged by the administration of UA, and only minimal effects on the immune response to BDV Ags were observed. These results indicate that the CNS inflammatory response to neurotropic virus infection is likely to be dependent upon the activity of ONOO(-) or its products on the blood-brain barrier.


Subject(s)
Blood-Brain Barrier/drug effects , Borna Disease/immunology , Borna disease virus/immunology , Brain/immunology , Chemotaxis, Leukocyte/physiology , Encephalitis, Viral/immunology , Free Radical Scavengers/therapeutic use , Neuroprotective Agents/therapeutic use , Peroxynitrous Acid/physiology , Tyrosine/analogs & derivatives , Uric Acid/therapeutic use , Acute Disease , Animals , Antigens, Viral/immunology , Borna Disease/pathology , Borna Disease/virology , Borna disease virus/physiology , Brain/metabolism , Brain/pathology , Brain/virology , Brain Chemistry/drug effects , Chemotaxis, Leukocyte/drug effects , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Female , Free Radical Scavengers/pharmacology , Free Radicals , Gene Expression Profiling , Immunocompetence , Inflammation , Lymphocyte Count , Nerve Tissue Proteins/analysis , Neurons/enzymology , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Oxidation-Reduction , Polymerase Chain Reaction , Rats , Rats, Inbred Lew , T-Lymphocyte Subsets/drug effects , Tyrosine/analysis , Uric Acid/pharmacology , Virus Replication/drug effects
10.
Mult Scler ; 7(3): 165-6, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11475439

ABSTRACT

Presence of nitrotyrosine in cells surrounding plaques indicates that peroxynitrite may be the cause of brain lesions in multiple sclerosis. Low levels of uric acid, a natural scavenger of peroxynitrite, were demonstrated in blood of patients with multiple sclerosis in comparison with control individuals. These observations were now extended to 132 sets of twins with one sibling affected by multiple sclerosis. In blood of both mono- and dizygotic twins the uric acid levels were lower in the twin with the disease than in the healthy twin.


Subject(s)
Multiple Sclerosis/blood , Uric Acid/blood , Female , Humans , Male , Twins, Dizygotic , Twins, Monozygotic
12.
J Immunol ; 165(11): 6511-8, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11086092

ABSTRACT

Uric acid (UA), a product of purine metabolism, is a known scavenger of peroxynitrite (ONOO(-)), which has been implicated in the pathogenesis of multiple sclerosis and experimental allergic encephalomyelitis (EAE). To determine whether the known therapeutic action of UA in EAE is mediated through its capacity to inactivate ONOO(-) or some other immunoregulatory phenomenon, the effects of UA on Ag presentation, T cell reactivity, Ab production, and evidence of CNS inflammation were assessed. The inclusion of physiological levels of UA in culture effectively inhibited ONOO(-)-mediated oxidation as well as tyrosine nitration, which has been associated with damage in EAE and multiple sclerosis, but had no inhibitory effect on the T cell-proliferative response to myelin basic protein (MBP) or on APC function. In addition, UA treatment was found to have no notable effect on the development of the immune response to MBP in vivo, as measured by the production of MBP-specific Ab and the induction of MBP-specific T cells. The appearance of cells expressing mRNA for inducible NO synthase in the circulation of MBP-immunized mice was also unaffected by UA treatment. However, in UA-treated animals, the blood-CNS barrier breakdown normally associated with EAE did not occur, and inducible NO synthase-positive cells most often failed to reach CNS tissue. These findings are consistent with the notion that UA is therapeutic in EAE by inactivating ONOO(-), or a related molecule, which is produced by activated monocytes and contributes to both enhanced blood-CNS barrier permeability as well as CNS tissue pathology.


Subject(s)
Blood-Brain Barrier/immunology , Cell Movement/immunology , Central Nervous System/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Free Radical Scavengers/pharmacology , Nitrates/metabolism , Uric Acid/pharmacology , Animals , Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Capillary Permeability/immunology , Cell Movement/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Free Radical Scavengers/administration & dosage , Free Radical Scavengers/metabolism , Injections, Intraperitoneal , Injections, Subcutaneous , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred Strains , Myelin Basic Protein/administration & dosage , Myelin Basic Protein/immunology , Nitrates/antagonists & inhibitors , Oxidation-Reduction , Uric Acid/administration & dosage , Uric Acid/metabolism
13.
Neurosci Lett ; 292(2): 137-41, 2000 Oct 06.
Article in English | MEDLINE | ID: mdl-10998568

ABSTRACT

Peroxynitrite (ONOO(-)), the product of nitric oxide (NO(radical)) and superoxide (O(2)(-radical)), is believed to be a major contributor to immunotoxicity when produced by activated cells expressing inducible nitric oxide synthase (iNOS). Uric acid (UA) is a natural scavenger of ONOO(-) that is present at high levels in the sera of humans and other higher order primates relative to most lower mammals. We have previously shown that UA treatment is therapeutic in experimental allergic encephalomyelitis (EAE), a rodent model of multiple sclerosis (MS). In this study we have examined the effect of UA therapy on the dynamics of the appearance of iNOS-positive cells in central nervous system (CNS) tissue of mice subjected to the stimuli that cause EAE. The results indicate that UA prevents activated monocytes from entering CNS tissue where they may contribute to the pathogenesis of MS and other CNS diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Monocytes/immunology , Myelin Basic Protein/immunology , Myelin Basic Protein/metabolism , Uric Acid/pharmacokinetics , Animals , Blood-Brain Barrier/physiology , Disease Models, Animal , Female , Free Radicals/metabolism , Gene Expression Regulation, Enzymologic/immunology , Immunization , Lipid Peroxidation/drug effects , Mice , Mice, Inbred Strains , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Nitrates/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/analysis
14.
FASEB J ; 14(5): 691-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10744626

ABSTRACT

Peroxynitrite (ONOO(-)), a toxic product of the free radicals nitric oxide and superoxide, has been implicated in the pathogenesis of CNS inflammatory diseases, including multiple sclerosis and its animal correlate experimental autoimmune encephalomyelitis (EAE). In this study we have assessed the mode of action of uric acid (UA), a purine metabolite and ONOO(-) scavenger, in the treatment of EAE. We show that if administered to mice before the onset of clinical EAE, UA interferes with the invasion of inflammatory cells into the CNS and prevents development of the disease. In mice with active EAE, exogenously administered UA penetrates the already compromised blood-CNS barrier, blocks ONOO(-)-mediated tyrosine nitration and apoptotic cell death in areas of inflammation in spinal cord tissues and promotes recovery of the animals. Moreover, UA treatment suppresses the enhanced blood-CNS barrier permeability characteristic of EAE. We postulate that UA acts at two levels in EAE: 1) by protecting the integrity of the blood-CNS barrier from ONOO(-)-induced permeability changes such that cell invasion and the resulting pathology is minimized; and 2) through a compromised blood-CNS barrier, by scavenging the ONOO(-) directly responsible for CNS tissue damage and death.


Subject(s)
Blood-Brain Barrier/drug effects , Free Radical Scavengers/pharmacology , Multiple Sclerosis/drug therapy , Nitrates/metabolism , Uric Acid/pharmacology , Animals , Apoptosis/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Mice , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Tyrosine/analogs & derivatives , Tyrosine/metabolism
15.
Proc Natl Acad Sci U S A ; 96(5): 2549-53, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10051680

ABSTRACT

Alfalfa mosaic virus (AlMV) coat protein is involved in systemic infection of host plants, and a specific mutation in this gene prevents the virus from moving into the upper uninoculated leaves. The coat protein also is required for different viral functions during early and late infection. To study the role of the coat protein in long-distance movement of AlMV independent of other vital functions during virus infection, we cloned the gene encoding the coat protein of AlMV into a tobacco mosaic virus (TMV)-based vector Av. This vector is deficient in long-distance movement and is limited to locally inoculated leaves because of the lack of native TMV coat protein. Expression of AlMV coat protein, directed by the subgenomic promoter of TMV coat protein in Av, supported systemic infection with the chimeric virus in Nicotiana benthamiana, Nicotiana tabacum MD609, and Spinacia oleracea. The host range of TMV was extended to include spinach as a permissive host. Here we report the alteration of a host range by incorporating genetic determinants from another virus.


Subject(s)
Alfalfa mosaic virus/genetics , Capsid/genetics , Tobacco Mosaic Virus/genetics , Alfalfa mosaic virus/metabolism , Amino Acid Sequence , Capsid/biosynthesis , Capsid/chemistry , Cloning, Molecular , Genome, Viral , Molecular Sequence Data , Plant Diseases , Plant Leaves , Plants, Toxic , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Spinacia oleracea/virology , Nicotiana/virology , Transcription, Genetic
16.
J Virol ; 73(1): 510-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9847357

ABSTRACT

The mouse-adapted rabies virus strain CVS-24 has stable variants, CVS-B2c and CVS-N2c, which differ greatly in their pathogenicity for normal adult mice and in their ability to infect nonneuronal cells. The glycoprotein (G protein), which has previously been implicated in rabies virus pathogenicity, shows substantial structural differences between these variants. Although prior studies have identified antigenic site III of the G protein as the major pathogenicity determinant, CVS-B2c and CVS-N2c do not vary at this site. The possibility that pathogenicity is inversely related to G protein expression levels is suggested by the finding that CVS-B2c, the less pathogenic variant, expresses at least fourfold-higher levels of G protein than CVS-N2c in infected neurons. Although there is some difference between CVS-B2c- and CVS-N2c-infected neurons in G protein mRNA expression levels, the differential expression of G protein appears to be largely determined by posttranslational mechanisms that affect G protein stability. Pulse-chase experiments indicated that the G protein of CVS-B2c is degraded more slowly than that of CVS-N2c. The accumulation of G protein correlated with the induction of programmed cell death in CVS-B2c-infected neurons. The extent of apoptosis was considerably lower in CVS-N2c-infected neurons, where G protein expression was minimal. While nucleoprotein (N protein) expression levels were similar in neurons infected with either variant, the transport of N protein into neuronal processes was strongly inhibited in CVS-B2c-infected cells. Thus, downregulation of G protein expression in neuronal cells evidently contributes to rabies virus pathogenesis by preventing apoptosis and the apparently associated failure of the axonal transport of N protein.


Subject(s)
Antigens, Viral , Apoptosis , Glycoproteins/physiology , Neurons/virology , Rabies virus/pathogenicity , Viral Envelope Proteins/physiology , Animals , Axonal Transport , Cells, Cultured , Glycoproteins/genetics , Mice , Nucleocapsid/genetics , Nucleocapsid/physiology , Nucleocapsid Proteins , RNA, Messenger/analysis , Rabies virus/metabolism , Viral Envelope Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 95(2): 675-80, 1998 Jan 20.
Article in English | MEDLINE | ID: mdl-9435251

ABSTRACT

Uric acid, the naturally occurring product of purine metabolism, is a strong peroxynitrite scavenger, as demonstrated by the capacity to bind peroxynitrite but not nitric oxide (NO) produced by lipopolysaccharide-stimulated cells of a mouse monocyte line. In this study, we used uric acid to treat experimental allergic encephalomyelitis (EAE) in the PLSJL strain of mice, which develop a chronic form of the disease with remissions and exacerbations. Uric acid administration was found to have strong therapeutic effects in a dose-dependent fashion. A regimen of four daily doses of 500 mg/kg uric acid was required to promote long-term survival regardless of whether treatment was initiated before or after the clinical symptoms of EAE had appeared. The requirement for multiple doses is likely to be caused by the rapid clearance of uric acid in mice which, unlike humans, metabolize uric acid a step further to allantoin. Uric acid treatment also was found to diminish clinical signs of a disease resembling EAE in interferon-gamma receptor knockout mice. A possible association between multiple sclerosis (MS), the disease on which EAE is modeled, and uric acid is supported by the finding that patients with MS have significantly lower levels of serum uric acid than controls. In addition, statistical evaluation of more than 20 million patient records for the incidence of MS and gout (hyperuricemic) revealed that the two diseases are almost mutually exclusive, raising the possibility that hyperuricemia may protect against MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis/drug therapy , Nitrates/metabolism , Uric Acid/administration & dosage , Animals , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Humans , Mice , Mice, Knockout , Multiple Sclerosis/metabolism , Uric Acid/blood
18.
Mol Med ; 3(5): 315-26, 1997 May.
Article in English | MEDLINE | ID: mdl-9205947

ABSTRACT

BACKGROUND: The production of nitric oxide by type II inducible nitric oxide synthase (type II NOS) gene is controlled at least in part by transcriptional activation. Although the murine and human type II NOS genes share significant sequence homology, they differ in the induction stimuli required for activation. MATERIALS AND METHODS: The A549 human and murine RAW 264.7 cell lines were cultured in the presence of inducers of the type II NOS gene and exposed to specific inhibitors of phosphatidyl choline-specific phospholipase C, NF-kappa B, and endocytosis, as well as to reagents that deplete stores of ATP or prevent the acidification of endosomes. The effect of these reagents on the induction of the type II NOS gene transcription, translation, and NO expression was studied using electromobility shift assays, Western blotting, and the detection of NO as nitrates, as appropriate. Additionally, the ability of the native human type II NOS NF-kappa B recognition sequence to bind NF-kappa B was compared with a concensus sequence and with a mutated oligomer. RESULTS: Type II NOS production by both human and mouse cells could be prevented by the addition of the specific inhibitor of phosphatidylcholine-specific phospholipase C, D609, and of agents that interfere with the activation of NF-kappa B. Both mouse and human cells also required acidic endosome formation and the production of 1,2-diacylglycerol for type II NOS expression. Additionally, the native human type II NOS NF-kappa B recognition sequence bound NF-kappa B with significantly less affinity than did the recognition sequence derived from the human immunoglobulin light-chain gene promoter. CONCLUSIONS: These experiments show that whereas mouse cells can be activated by lipopolysaccharide to produce nitric oxide, and human cells require activation by a mixture of cytokines to produce nitric oxide, the intracellular activation pathway following receptor binding of these heterologous stimuli is shared. Additionally, NF-kappa B activation is necessary but not sufficient for inducible nitric oxide synthase production in human cells, in contrast to murine cells in which it serves as a complete inducer.


Subject(s)
NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Transcriptional Activation , Type C Phospholipases/metabolism , Animals , Antioxidants/pharmacology , Blotting, Western , Bridged-Ring Compounds/pharmacology , Cell Line , Cytokines/metabolism , Epithelium , Gene Expression , Humans , Lipid Metabolism , Lung/cytology , Lung/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , NF-kappa B/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Norbornanes , Phosphatidylcholines/metabolism , Phosphodiesterase Inhibitors/pharmacology , RNA, Messenger , RNA-Binding Proteins/metabolism , Ribonucleases , Thiocarbamates , Thiones/pharmacology , Type C Phospholipases/antagonists & inhibitors
19.
J Neurovirol ; 3(2): 174-8, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9111180

ABSTRACT

The presence of antibodies reactive with Borna disease virus (BDV) in the sera of some patients with certain psychiatric illnesses has been taken as evidence that this veterinary neurotrophic virus may occasionally infect and cause psychiatric disorders in humans. In this paper, we report the results of our studies concerning the detection of BDV-specific RNA in blood cells from patients with psychiatric diseases. Contrary to the results obtained by others, we have found no evidence for the presence of BDV-RNA in such cells. Prior work with BDV sequences in the assay environment, together with the exquisite sensitivity of RT-PCR, may account for the sporadic appearance of false positive evidence that BDV-specific RNA is present in human blood cells.


Subject(s)
Borna Disease/blood , Borna disease virus/isolation & purification , Mental Disorders/virology , Adult , Animals , Borna Disease/complications , Borna Disease/diagnosis , Cohort Studies , False Positive Reactions , Female , Humans , Leukocytes/virology , Male , Mental Disorders/blood , Mental Disorders/etiology , Middle Aged , Polymerase Chain Reaction , RNA, Viral/blood , Rabbits , Schizophrenia/blood , Schizophrenia/etiology , Schizophrenia/virology , Sensitivity and Specificity
20.
Mol Med ; 2(2): 226-35, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8726465

ABSTRACT

BACKGROUND: Nitric oxide has a wide variety of homeostatic and pathological effects. Control of the production of nitric oxide by the inducible form of the enzyme resides in the 5' promoter region of the gene. Although control of the murine isoform has been investigated, little is known about the functional aspects of the human analog. MATERIALS AND METHODS: A 3.9-kb 5' nontranslated region of the human gene was cloned, sequenced, and several reporter constructs prepared. The promoter-reporter constructs were transfected into human or murine monocytoid cells and reporter expression quantified following cytokine activation of the cells. The production of nitric oxide was also monitored. RESULTS: Although a murine promoter-reporter functioned efficiently in both human and mouse cells, the human constructs functioned only in human cells. The activity of the mouse construct increased progressively with the addition of activating cytokines, but the human promoter-reporter did not. Although interleukin 1 beta drove expression of the human inducible nitric oxide synthase reporter, actual expression of nitric oxide required both interleukin 1 beta and interferon-gamma. CONCLUSIONS: The data indicate that despite the significant homology between the human and mouse inducible nitric oxide synthase promoter sequence, control of the two genes is quite different. In addition to being more efficient in promoter activity, the murine promoter responds increasingly to cytokines that are not effective for the human analog. It is also apparent that human inducible nitric oxide synthase is controlled at both the level of transcription and post-translationally.


Subject(s)
Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Promoter Regions, Genetic , Transfection , Animals , Base Sequence , Cell Line , Consensus Sequence , DNA Primers , Female , Genes, Reporter , Genomic Library , Humans , Luciferases/biosynthesis , Macrophages , Mice , Molecular Sequence Data , Placenta/enzymology , Polymerase Chain Reaction/methods , Pregnancy , Recombinant Fusion Proteins/biosynthesis , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...