Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Osteoarthritis Cartilage ; 10(11): 838-44, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12435327

ABSTRACT

Objective To assess the correlation between changes in spin-lattice relaxation in the rotating frame (T(1rho)) and proteoglycan (PG) loss from bovine articular cartilage and to demonstrate the feasibility of performing T(1rho) MR imaging on a 1.5T clinical scanner. Design MR relaxation times (T(1rho), T(2) and T(1)) were measured from excised cartilage plugs (N=3) before and after two sequential digestions with trypsin on a 2T whole-body magnet. Proteoglycan and collagen loss induced by the trypsin digestion was measured using standard biochemical techniques. The correlation between changes in relaxation times and PG loss were tested with regression analysis. T(1rho) MRI was also performed on a clinical 1.5T MRI system to determine whether the spatial distribution of PG loss could be detected. The MRI results were compared with histology sections of native and PG-depleted tissue. Results Increase in T(1rho) relaxation times correlated with PG loss (R(2)=0.81). T(1rho) measurements alone were indicative of PG loss (R(2)=0.8), the addition of T1 and T2 data into the statistical model did not improve the correlation substantially (R(2)=0.83). T(1rho)-weighted imaging demonstrated a hyperintense lamina at the articular surface of the digested tissue, which was subjected to trypsin digestion that correlated with a superficial zone of PG loss observed on histological sections. Conclusion The results of this study demonstrate that T(1rho) relaxation changes are correlated with PG loss in vitro. Furthermore, T(1rho) measurements alone can be used to indicate PG loss data. T(1rho) MRI may thus be developed into a useful adjunct to existing techniques for the evaluation of cartilage disease.


Subject(s)
Cartilage, Articular/metabolism , Magnetic Resonance Imaging/methods , Proteoglycans/metabolism , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cattle , Collagen/analysis , Hydroxyproline/analysis , Patella/metabolism , Patella/pathology , Time Factors , Trypsin/pharmacology
2.
J Infect Dis ; 184(7): 809-16, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11550123

ABSTRACT

West Nile virus (WNV) is a vectorborne pathogen that induces brain inflammation and death. Recently, confirmed cases of infection and deaths have occurred in the United States Mid-Atlantic region. In this study, a DNA vaccine encoding the WNV capsid protein was constructed, and the in vivo immune responses generated were investigated in DNA vaccine-immunized mice. Antigen-specific humoral and cellular immune responses were observed, including a potent induction of antigen-specific Th1 and cytotoxic T lymphocyte responses. Strong induction of Th1-type immune responses included high levels of antigen-specific elaboration of the Th1-type cytokines interferon-gamma and interleukin-2 and beta-chemokines RANTES (regulated upon activation, normal T cell-expressed and secreted) and macrophage inflammatory protein-1beta. Dramatic infiltration of CD4 and CD8 T cells and macrophages also was observed at the muscle injection site. These results support the potential utility of this method as a tool for developing immunization strategies for WNV and other emerging pathogens.


Subject(s)
T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Vaccines, DNA/administration & dosage , Viral Vaccines/administration & dosage , West Nile Fever/prevention & control , West Nile virus/immunology , Animals , Antigens, Viral/immunology , Capsid/immunology , Cytokines/analysis , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Muscles/immunology , West Nile virus/isolation & purification
3.
Radiology ; 220(3): 822-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11526288

ABSTRACT

A fast spin-echo sequence weighted with a time constant that defines the magnetic relaxation of spins under the influence of a radio-frequency field (T1(rho)) was used in six subjects to measure magnetic resonance (MR) relaxation times in the knee joint with a 1.5-T MR imager. A quantitative comparison of T2- and T1(rho)-weighted MR images was also performed. Substantial T1(rho) dispersion was demonstrated in human articular cartilage, but muscle did not demonstrate much dispersion. T1(rho)-weighted images depicted a chondral lesion with 25% better signal-difference-to-noise ratios than comparable T2-weighted images. This technique may depict cartilage and muscular abnormalities.


Subject(s)
Knee Joint , Magnetic Resonance Imaging/methods , Adult , Cartilage, Articular , Female , Humans , Joint Diseases/diagnosis , Male , Muscle, Skeletal , Pain/diagnosis
4.
Arthritis Rheum ; 44(5): 1089-95, 2001 May.
Article in English | MEDLINE | ID: mdl-11352240

ABSTRACT

OBJECTIVE: To determine the metabolic fate of glucosamine (GlcN) in intact articular cartilage tissue. METHODS: Intact articular cartilage explants were cultured for up to 13 days in Dulbecco's modified Eagle's medium supplemented with 1) 1-13C-labeled GlcN, 2) 1-13C-labeled glucose (Glc), or 3) no labeling. Every 3-4 days, samples were removed and frozen in liquid nitrogen for carbon-13 magnetic resonance spectroscopic (MRS) analysis. The metabolic products of the labeled precursors were determined from the MRS data based on resonance positions and comparison with known standards and published values. RESULTS: GlcN was taken up by the chondrocytes and incorporated selectively into the hexosamine, but not the hexuronic acid, components of the glycosaminoglycan chains of articular cartilage proteoglycan. The data also demonstrated that GlcN is the substrate of choice for the galactosamine moieties of the chondroitin sulfates, incorporating at levels 300% higher than with an equivalent amount of labeled Glc. CONCLUSION: The results indicate that GlcN facilitates the production of proteoglycan components that are synthesized through the hexosamine biochemical pathway.


Subject(s)
Cartilage, Articular/metabolism , Chondroitin Sulfates/metabolism , Galactosamine/metabolism , Glucosamine/pharmacokinetics , Animals , Carbon Isotopes , Cartilage, Articular/cytology , Cattle , Cells, Cultured , Magnetic Resonance Spectroscopy
5.
DNA Cell Biol ; 20(11): 723-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11788050

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by overproduction of beta-amyloid (Abeta), which is formed from amyloid precursor protein (APP), with the subsequent pathologic deposition of Abeta in regions of the brain important for memory and cognition. Recently, vaccination of murine models of AD that exhibit Abeta deposition has halted or delayed the usual progression of the pathology of AD. Our group has demonstrated that vaccination of a doubly transgenic mouse model (expressing mutant APP and presenilin-1) with the Abeta 1-42 peptide protects these mice from the memory deficits they would ordinarily develop. This report further characterizes the Abeta 1-42 peptide vaccine in mice. Anti-Abeta response time course analysis indicated that at least three vaccinations (each 100 microg) were necessary to elicit a significant anti-Abeta titer. Subsequent vaccinations resulted in half-maximal antibody titers of at least 10,000, and these titers were maintained for at least 5 months after the final boost. Peptide binding competition studies indicated that the highest humoral responses are generated against the N terminus of the Abeta peptide. Also, measurement of specific murine Ig isotypes in Abeta-vaccinated mice demonstrated a predominant IgG(1) and IgG(2b) response, suggesting a type 2 (Th2) T-helper cell immune response, which drives humoral immunity. Finally, lymphocyte proliferation assay experiments using Abeta peptides and splenocytes from vaccinated mice demonstrated that the vaccine specifically stimulates T-cell epitopes present within the Abeta peptide.


Subject(s)
Amyloid beta-Peptides/administration & dosage , Antibody Formation , Antibody Specificity , Peptide Fragments/administration & dosage , Vaccines/administration & dosage , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Immunoglobulins/classification , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/immunology , Sequence Homology, Amino Acid
6.
J Leukoc Biol ; 68(3): 366-72, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10985253

ABSTRACT

The HIV-1 vpr gene encodes a 14-kDa virion-packaged protein that has been implicated in viral pathogenesis. Vpr exhibits profound effects on human primary cells influencing proliferation, differentiation, apoptosis, and cytokine production, in part through NF-kappaB-mediated transcription. NF-kappaB, a potent transcription factor, activates many proinflammatory cytokines/chemokines upon infection. Here, we analyzed the effect of extracellular Vpr as well as the virion-associated Vpr on beta chemokines (MIP-1alpha, MIP-1beta, and RANTES) production in human macrophages and primary lymphocytes (PBLs). Macrophages and PBLs exposed to HIV-1 vpr+ viruses or to recombinant Vpr protein produced significantly less beta chemokines compared with cells infected with HIV-1 vpr-viruses or irrelevant control protein (Gag)-exposed cells. These results suggest that a Vpr-mediated increase in virus replication could be in part through down-regulation of chemokine production.


Subject(s)
Chemokines, CC/biosynthesis , Gene Products, vpr/physiology , HIV-1/genetics , Lymphocytes/metabolism , Macrophages/metabolism , Chemokines, CC/genetics , Gene Expression Regulation, Viral/physiology , Gene Products, vpr/genetics , Gene Products, vpr/pharmacology , Genes, vpr , HIV-1/physiology , HeLa Cells , Humans , Lymphocytes/virology , Macrophages/virology , Recombinant Proteins/pharmacology , Virion/genetics , Virion/metabolism , Virus Replication/physiology , vpr Gene Products, Human Immunodeficiency Virus
7.
Osteoarthritis Cartilage ; 8(4): 288-93, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10903883

ABSTRACT

OBJECTIVE: The purpose of this work was to evaluate the results from sodium and proton magnetic resonance imaging (MRI) in detecting small changes in proteoglycan (PG) content in bovine articular cartilage specimens. DESIGN: Articular cartilage from 15 specimens of bovine patellae were subjected to partial PG depletion with different concentrations of trypsin for 30 min. Sodium and proton MR images of the intact specimen were obtained on a 4T GE clinical MRI system. Two custom-built 7 cm-diameter solenoid coils tuned to proton and sodium frequencies were employed. Fast gradient echo and spin echo imaging sequences were used to determine sodium density, proton density and proton relaxation times (T(1)and T(2)) of the specimens. Spectrophotometric assay was performed after MRI to determine PG concentrations of the cartilage specimens. RESULTS: The sodium signal change correlated well with the observed PG loss (R(2)=0.85, P< 0.01) whereas the proton signal change was inconsistent (R(2)=0.10, P< 0.8). The change in proton T(1)and T(2)between the two regions did not correlate with PG loss (R(2)=0. 07 and R(2)=0.06, respectively). CONCLUSIONS: Results from these studies demonstrate that sodium MRI is both sensitive and specific in detecting small changes in PG concentration, whereas proton density and relaxation properties are not sensitive to small changes in PG content.


Subject(s)
Cartilage, Articular/anatomy & histology , Proteoglycans/analysis , Animals , Cartilage, Articular/drug effects , Cattle , Magnetic Resonance Imaging/methods , Proteoglycans/drug effects , Sensitivity and Specificity , Spectrophotometry , Trypsin/pharmacology
8.
AIDS ; 14(1): 1-9, 2000 Jan 07.
Article in English | MEDLINE | ID: mdl-10714562

ABSTRACT

OBJECTIVE: To develop an HIV-1 accessory gene immunogen using a DNA vaccine approach. METHODS: HIV-1 accessory genes vif, vpu and nef were modified to express under the control of a single promoter with cellular proteolytic cleavage sites between the coding sequences (VVN-P). Immune responses induced by these constructs were evaluated in mice. RESULTS: DNA vaccine construct (pVVN-P) expressing Vif, Vpu and Nef was processed and the fusion protein was cleaved appropriately. Vif, Vpu and Nef as a fusion protein with proteolytic cleavage sites (VVN-P) is able to induce a significant level of cellular immune responses. We also observed that accessory genes Vif, Vpu and Nef (VVN-P) induced an effective T helper 1 proliferative response measured by cytokine production. Furthermore, expression cassette pVVN-P was able to induce cytotoxic T lymphocyte (CTL) responses against diverse HIV-1 viruses in infected target cells. CONCLUSION: We conclude that cell-mediated immune responses induced by accessory gene constructs from clade B may have a broader recognition of divergent HIV-1 viruses and should be further examined for both prophylactic and therapeutic vaccination schemes against HIV-1.


Subject(s)
HIV-1/immunology , Vaccines, DNA/immunology , 3T3 Cells , Animals , Blotting, Western , Cell Division/immunology , Cells, Cultured , Cytokines/biosynthesis , Cytotoxicity Tests, Immunologic , Fluorescent Antibody Technique , Gene Products, nef/genetics , Gene Products, nef/immunology , Gene Products, nef/metabolism , Gene Products, vif/genetics , Gene Products, vif/immunology , Gene Products, vif/metabolism , HIV-1/genetics , HIV-1/isolation & purification , HeLa Cells , Human Immunodeficiency Virus Proteins , Humans , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Transfection , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/metabolism , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/immunology , Viral Regulatory and Accessory Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus , vif Gene Products, Human Immunodeficiency Virus
9.
Nat Med ; 3(10): 1117-23, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9334723

ABSTRACT

The HIV-1 accessory gene product Vpr can influence viral pathogenesis by affecting viral replication as well as host cell transcription and proliferation. We have investigated the effects of Vpr on host cell activation and confirm that it influences cellular proliferation. However, we have also found that Vpr modulates T-cell receptor (TCR)-triggered apoptosis in a manner similar to that of glucocorticoids. In the absence of TCR-mediated activation, Vpr induces apoptosis whereas in its presence, Vpr interrupts the expected induction of apoptosis. This regulation of apoptosis is linked to Vpr suppression of NF-kappa B activity via the induction of I kappa B, an inhibitor of NF-kappa B. Further, Vpr suppresses expression of IL-2, IL-10, IL-12, TNF alpha and IL-4, all of which are NF-kappa B-dependent. The effects of Vpr could be reversed by RU486. Our finding that Vpr can regulate NF-kappa B supports the hypothesis that some aspects of viral pathogenesis are the consequence of cell dysregulation by Vpr.


Subject(s)
Apoptosis/drug effects , Cytokines/biosynthesis , Gene Products, vpr/pharmacology , HIV-1/immunology , Lymphocyte Activation/drug effects , Lymphocytes/immunology , NF-kappa B/metabolism , Animals , Cell Line , Cells, Cultured , Dexamethasone/pharmacology , Gene Products, vpr/biosynthesis , Humans , Hydrocortisone/pharmacology , Interleukins/biosynthesis , Lymphocytes/drug effects , Lymphocytes/virology , Phytohemagglutinins , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Spodoptera , Transfection , Tumor Necrosis Factor-alpha/biosynthesis , vpr Gene Products, Human Immunodeficiency Virus
10.
J Leukoc Biol ; 62(1): 93-9, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9225999

ABSTRACT

Among the putative accessory genes of HIV-1, the 96-amino-acid virion-associated vpr gene product has been described to have several novel biological activities. These include cytoplasmic-to-nuclear translocation, which empowers HIV to infect and replicate in non-dividing cells and to increase viral replication, particularly in macrophages. Along with these viral effects, we found that HIV-1 Vpr induces dramatic biological changes in the target cells of HIV infection, including induction of changes in transcriptional patterns, morphological changes, and complete inhibition of proliferation, which collectively was termed differentiation. These changes occur in the absence of other viral gene products, suggesting that Vpr mediates its proviral effects partially or perhaps solely through modulation of the state of the target cell rather than directly on the virus. The inhibition of proliferation in T cell lines has been extended by several groups to demonstrate that the inhibition of proliferation is through G2 cell cycle arrest, further supporting the idea that Vpr acts directly on cellular targets. We have recently described a role for Vpr in modulating the glucocorticoid pathway, which is involved in the regulation of the state of the cell, in cytoplasmic-to-nuclear translocation, and in modulation of host cell transcription. It is important to note that certain anti-glucocorticoid compounds modulate Vpr activity in vitro. These results support the idea that the host cell contains specific receptor molecule(s) through which Vpr mediates its activity. Consequently, Vpr represents a unique target for anti-HIV drug development and has significance for HIV-1 disease progression.


Subject(s)
Gene Products, vpr/metabolism , HIV-1/physiology , Monocytes/virology , T-Lymphocytes/virology , Virus Replication , Amino Acid Sequence , Cell Cycle/drug effects , Cell Differentiation , Cells, Cultured , Gene Products, vpr/chemistry , Gene Products, vpr/pharmacology , Glucocorticoids/metabolism , Humans , Molecular Sequence Data , Peptide Fragments/pharmacology , Proviruses/physiology , Receptors, HIV/physiology , T-Lymphocytes/cytology , vpr Gene Products, Human Immunodeficiency Virus
11.
AIDS Res Hum Retroviruses ; 13(18): 1561-72, 1997 Dec 10.
Article in English | MEDLINE | ID: mdl-9430248

ABSTRACT

We have previously reported on the generation of specific functional immune responses after inoculation of animals with expression vectors encoding HIV-1 genes. This article provides the details of the first application of this new technology to induce immune responses against HIV-2. This virus is molecularly and serologically distinct from HIV-1 and is in fact more closely related to the simian immunodeficiency virus (SIV). Anti-HIV-2 and SIV antibodies were induced in mice of three different haplotypes following a single intramuscular inoculation with an HIV-2/ROD envelope glycoprotein expression vector (pcEnv-2). Boosting of animals with pcEnv-2 induced both anti-HIV-2 neutralizing antibodies and T cell-proliferative responses against HIV-2 and SIVmac proteins. We compared the humoral and cellular immune responses of mice injected with pcEnv-2 and then boosted with either the homologous DNA construct or a recombinant Env protein. Animals boosted with pcEnv-2 generated B and T cell immune responses as strong as those of mice boosted with recombinant gp140 protein in adjuvant. Finally, cellular immune responses were significantly increased with the coadministration of pcEnv-2 and a plasmid expressing interleukin 12. We therefore conclude that DNA plasmid inoculation induces cross-reactive anti-HIV-2 and anti-SIVmac immune responses in mice. This technology should be further investigated as a potential vaccine component for this human pathogen.


Subject(s)
AIDS Vaccines/immunology , DNA, Viral/immunology , HIV Antibodies/immunology , HIV-2/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, DNA/immunology , Vaccines, Synthetic/immunology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cross Reactions , Female , HIV Antibodies/biosynthesis , Humans , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmids , T-Lymphocytes/immunology , Tumor Cells, Cultured
12.
Immunol Res ; 13(2-3): 154-62, 1994.
Article in English | MEDLINE | ID: mdl-7775806

ABSTRACT

There are a number of rheumatologic manifestations of human retroviral infections associated with human immunodeficiency virus type I (HIV-I) and the human T-cell leukemia virus type I (HTLV-I) including arthritis, Sjøgren's syndrome-like symptoms as well as other varied autoimmune phenomena. Infection with HTLV-1 may be directly involved in the etiology and/or pathogenesis of an arthritic condition similar to rheumatoid arthritis. We have been characterizing a new vaccination strategy against human retroviral infections, designated DNA inoculation. This procedure involves the intramuscular injection of DNA plasmids which express specific human retroviral antigens. This technique results in the development of humoral and cellular immune responses against these proteins. Specifically, this method has been successfully used to develop immune responses against HIV-I and HTLV-I. The availability of rat and rabbit infection models for HTLV-I, coupled with the successful development of immune responses in these animals after DNA inoculation with an HTLV-I envelope expressing plasmid, will allow the efficacy of this vaccination technique to be evaluated with protection against in vivo viral challenge as an endpoint.


Subject(s)
DNA, Viral/therapeutic use , HIV Infections/prevention & control , HTLV-I Infections/prevention & control , Viral Vaccines/genetics , Animals , HIV Infections/complications , HTLV-I Infections/complications , Humans , Rheumatic Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL