Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
Mucosal Immunol ; 11(1): 186-198, 2018 01.
Article in English | MEDLINE | ID: mdl-28378805

ABSTRACT

The CD103+ subset of lung-migratory dendritic cells (DCs) plays an important role in the generation of CD8+ T cell responses following respiratory infection. Here, we demonstrate that the dependence on CD103+ DCs for stimulation of RSV-specific T cells is both epitope and age-dependent. CD103+ DCs in neonatal mice develop two phenotypically and functionally distinct populations following respiratory infection. Neonatal CD103+ DCs expressing low levels of CD103 (CD103lo DCs) and other lineage and maturation markers including costimulatory molecules are phenotypically immature and functionally limited. CD103lo DCs sorted from infected neonates were unable to stimulate cells of the KdM282-90 specificity, which are potently stimulated by CD103hi DCs sorted from the same animals. These data suggest that the delayed maturation of CD103+ DCs in the neonate limits the KdM282-90-specific response and explain the distinct CD8+ T cell response hierarchy displayed in neonatal mice that differs from the hierarchy seen in adult mice. These findings have implications for the development of early-life vaccines, where the promotion of responses with less age bias may prove advantageous. Alternately, specific approaches may be used to enhance the maturation and function of the CD103lo DC population in neonates to promote more adult-like T cell responses.


Subject(s)
Dendritic Cells/immunology , Lung/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes/immunology , Animals , Animals, Newborn , Antigens, CD/metabolism , Antigens, Viral/immunology , Cell Differentiation , Cell Lineage , Cell Movement , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , Female , Humans , Integrin alpha Chains/metabolism , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype
2.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28468888

ABSTRACT

Respiratory syncytial virus (RSV) infections remain a major cause of respiratory disease and hospitalizations among infants. Infection recurs frequently and establishes a weak and short-lived immunity. To date, RSV immunoprophylaxis and vaccine research is mainly focused on the RSV fusion (F) protein, but a vaccine remains elusive. The RSV F protein is a highly conserved surface glycoprotein and is the main target of neutralizing antibodies induced by natural infection. Here, we analyzed an internalization process of antigen-antibody complexes after binding of RSV-specific antibodies to RSV antigens expressed on the surface of infected cells. The RSV F protein and attachment (G) protein were found to be internalized in both infected and transfected cells after the addition of either RSV-specific polyclonal antibodies (PAbs) or RSV glycoprotein-specific monoclonal antibodies (MAbs), as determined by indirect immunofluorescence staining and flow-cytometric analysis. Internalization experiments with different cell lines, well-differentiated primary bronchial epithelial cells (WD-PBECs), and RSV isolates suggest that antibody internalization can be considered a general feature of RSV. More specifically for RSV F, the mechanism of internalization was shown to be clathrin dependent. All RSV F-targeted MAbs tested, regardless of their epitopes, induced internalization of RSV F. No differences could be observed between the different MAbs, indicating that RSV F internalization was epitope independent. Since this process can be either antiviral, by affecting virus assembly and production, or beneficial for the virus, by limiting the efficacy of antibodies and effector mechanism, further research is required to determine the extent to which this occurs in vivo and how this might impact RSV replication.IMPORTANCE Current research into the development of new immunoprophylaxis and vaccines is mainly focused on the RSV F protein since, among others, RSV F-specific antibodies are able to protect infants from severe disease, if administered prophylactically. However, antibody responses established after natural RSV infections are poorly protective against reinfection, and high levels of antibodies do not always correlate with protection. Therefore, RSV might be capable of interfering, at least partially, with antibody-induced neutralization. In this study, a process through which surface-expressed RSV F proteins are internalized after interaction with RSV-specific antibodies is described. One the one hand, this antigen-antibody complex internalization could result in an antiviral effect, since it may interfere with virus particle formation and virus production. On the other hand, this mechanism may also reduce the efficacy of antibody-mediated effector mechanisms toward infected cells.


Subject(s)
Antibodies, Viral/metabolism , Endocytosis , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/metabolism , Antigen-Antibody Complex/metabolism , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans
3.
Mucosal Immunol ; 10(2): 545-554, 2017 03.
Article in English | MEDLINE | ID: mdl-27220815

ABSTRACT

Cytomegalovirus vectors are promising delivery vehicles for vaccine strategies that aim to elicit effector CD8+ T cells. To determine how the route of immunization affects CD8+ T-cell responses in the lungs of mice vaccinated with a murine cytomegalovirus vector expressing the respiratory syncytial virus matrix (M) protein, we infected CB6F1 mice via the intranasal or intraperitoneal route and evaluated the M-specific CD8+ T-cell response at early and late time points. We found that intranasal vaccination generated robust and durable tissue-resident effector and effector memory CD8+ T-cell populations that were undetectable after intraperitoneal vaccination. The generation of these antigen-experienced cells by intranasal vaccination resulted in earlier T-cell responses, interferon gamma secretion, and viral clearance after respiratory syncytial virus challenge. Collectively, these findings validate a novel approach to vaccination that emphasizes the route of delivery as a key determinant of immune priming at the site of vulnerability.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Lung/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Viral Matrix Proteins/immunology , Viral Vaccines/immunology , Administration, Intranasal , Animals , CD8-Positive T-Lymphocytes/virology , Cell Line , Cytomegalovirus/genetics , Female , Genetic Vectors , Immunologic Memory , Injections, Intraperitoneal , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred Strains , Respiratory Syncytial Virus Infections/prevention & control , Vaccination , Viral Load , Viral Matrix Proteins/genetics
4.
Clin Exp Immunol ; 182(3): 289-301, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26332605

ABSTRACT

VRC-HIVMAB060-00-AB (VRC01) is a broadly neutralizing HIV-1 monoclonal antibody (mAb) isolated from the B cells of an HIV-infected patient. It is directed against the HIV-1 CD4 binding site and is capable of potently neutralizing the majority of diverse HIV-1 strains. This Phase I dose-escalation study in healthy adults was conducted at the National Institutes of Health (NIH) Clinical Center (Bethesda, MD, USA). Primary objectives were the safety, tolerability and pharmacokinetics (PK) of VRC01 intravenous (i.v.) infusion at 5, 20 or 40 mg/kg, given either once (20 mg/kg) or twice 28 days apart (all doses), and of subcutaneous (s.c.) delivery at 5 mg/kg compared to s.c. placebo given twice, 28 days apart. Cumulatively, 28 subjects received 43 VRC01 and nine received placebo administrations. There were no serious adverse events or dose-limiting toxicities. Mean 28-day serum trough concentrations after the first infusion were 35 and 57 µg/ml for groups infused with 20 mg/kg (n = 8) and 40 mg/kg (n = 5) doses, respectively. Mean 28-day trough concentrations after the second infusion were 56 and 89 µg/ml for the same two doses. Over the 5-40 mg/kg i.v. dose range (n = 18), the clearance was 0.016 l/h and terminal half-life was 15 days. After infusion VRC01 retained expected neutralizing activity in serum, and anti-VRC01 antibody responses were not detected. The human monoclonal antibody (mAb) VRC01 was well tolerated when delivered i.v. or s.c. The mAb demonstrated expected half-life and pharmacokinetics for a human immunoglobulin G. The safety and PK results support and inform VRC01 dosing schedules for planning HIV-1 prevention efficacy studies.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , HIV Antibodies , HIV Infections , HIV-1 , Adolescent , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/adverse effects , Broadly Neutralizing Antibodies , Dose-Response Relationship, Drug , Female , HIV Antibodies/administration & dosage , HIV Antibodies/adverse effects , HIV Infections/blood , HIV Infections/drug therapy , Half-Life , Humans , Male , Middle Aged
5.
Vaccine ; 31 Suppl 2: B209-15, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23598484

ABSTRACT

Although RSV has been a high priority for vaccine development, efforts to develop a safe and effective vaccine have yet to lead to a licensed product. Clinical and epidemiologic features of RSV disease suggest there are at least 4 distinct target populations for vaccines, the RSV naïve young infant, the RSV naïve child ≥ 6 months of age, pregnant women (to provide passive protection to newborns), and the elderly. These target populations raise different safety and efficacy concerns and may require different vaccination strategies. The highest priority target population is the RSV naïve child. The occurrence of serious adverse events associated with the first vaccine candidate for young children, formalin inactivated RSV (FI-RSV), has focused vaccine development for the young RSV naïve child on live virus vaccines. Enhanced disease is not a concern for persons previously primed by a live virus infection. A variety of live-attenuated viruses have been developed with none yet achieving licensure. New live-attenuated RSV vaccines are being developed and evaluated that maybe sufficiently safe and efficacious to move to licensure. A variety of subunit vaccines are being developed and evaluated primarily for adults in whom enhanced disease is not a concern. An attenuated parainfluenza virus 3 vector expressing the RSV F protein was evaluated in RSV naïve children. Most of these candidate vaccines have used the RSV F protein in various vaccine platforms including virus-like particles, nanoparticles, formulated with adjuvants, and expressed by DNA or virus vectors. The other surface glycoprotein, the G protein, has also been used in candidate vaccines. We now have tools to make and evaluate a wide range of promising vaccines. Costly clinical trials in the target population are needed to evaluate and select candidate vaccines for advancement to efficacy trials. Better data on RSV-associated mortality in developing countries, better estimates of the risk of long term sequelae such as wheezing after infection, better measures of protection in target populations, and data on the costs and benefits of vaccines for target populations are needed to support and justify funding this process. Addressing these challenges and needs should improve the efficiency and speed of achieving a safe and effective, licensed RSV vaccine.


Subject(s)
Biomedical Research/trends , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/therapeutic use , Aged , Child , Female , Humans , Infant , Pregnancy , Respiratory Syncytial Virus Vaccines/adverse effects , Vaccines, Attenuated/therapeutic use , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/therapeutic use , Vaccines, Subunit/therapeutic use , Viral Fusion Proteins/immunology
6.
Clin Vaccine Immunol ; 19(11): 1792-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22956656

ABSTRACT

Avian influenza virus causes outbreaks in domestic and wild birds around the world, and sporadic human infections have been reported. A DNA vaccine encoding hemagglutinin (HA) protein from the A/Indonesia/5/05 (H5N1) strain was initially tested in two randomized phase I clinical studies. Vaccine Research Center study 304 (VRC 304) was a double-blinded study with 45 subjects randomized to placebo, 1 mg of vaccine, or 4 mg of vaccine treatment groups (n = 15/group) by intramuscular (i.m.) Biojector injection. VRC 305 was an open-label study to evaluate route, with 44 subjects randomized to intradermal (i.d.) injections of 0.5 mg by needle/syringe or by Biojector or 1 mg delivered as two 0.5-mg Biojector injections in the same deltoid or as 0.5 mg in each deltoid (n = 11/group). Injections were administered at weeks 0, 4, and 8 in both studies. Antibody responses to H5 were assessed by hemagglutination inhibition (HAI) assay, enzyme-linked immunosorbent assay (ELISA), and neutralization assay, and the H5 T cell responses were assessed by enzyme-linked immunospot and intracellular cytokine staining assays. There were no vaccine-related serious adverse events, and the vaccine was well tolerated in all groups. At 1 mg, i.d. vaccination compared to i.m. vaccination induced a greater frequency and magnitude of response by ELISA, but there were no significant differences in the frequency or magnitude of response between the i.d. and i.m. routes in the HAI or neutralization assays. T cell responses were more common in subjects who received the 1- or 4-mg dose i.m. These studies demonstrated that the DNA vaccine encoding H5 is safe and immunogenic and served to define the proper dose and route for further studies. The i.d. injection route did not offer a significant advantage over the i.m. route, and no difference was detected by delivery to one site versus splitting the dose between two sites for i.d. vaccine administration. The 4-mg dose (i.m) was further investigated in prime-boost regimens.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Adult , Antibodies, Viral/blood , Cytokines/metabolism , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Female , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza Vaccines/genetics , Injections, Intradermal , Injections, Intramuscular , Male , Middle Aged , Neutralization Tests , Placebos/administration & dosage , T-Lymphocytes/immunology , Vaccines, DNA/genetics , Young Adult
7.
Vaccine ; 29(2): 304-13, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21034824

ABSTRACT

Ebola virus causes irregular outbreaks of severe hemorrhagic fever in equatorial Africa. Case mortality remains high; there is no effective treatment and outbreaks are sporadic and unpredictable. Studies of Ebola virus vaccine platforms in non-human primates have established that the induction of protective immunity is possible and safety and human immunogenicity has been demonstrated in a previous Phase I clinical trial of a 1st generation Ebola DNA vaccine. We now report the safety and immunogenicity of a recombinant adenovirus serotype 5 (rAd5) vaccine encoding the envelope glycoprotein (GP) from the Zaire and Sudan Ebola virus species, in a randomized, placebo-controlled, double-blinded, dose escalation, Phase I human study. Thirty-one healthy adults received vaccine at 2×10(9) (n=12), or 2×10(10) (n=11) viral particles or placebo (n=8) as an intramuscular injection. Antibody responses were assessed by ELISA and neutralizing assays; and T cell responses were assessed by ELISpot and intracellular cytokine staining assays. This recombinant Ebola virus vaccine was safe and subjects developed antigen specific humoral and cellular immune responses.


Subject(s)
Adenoviruses, Human/genetics , Ebola Vaccines/immunology , Genetic Vectors , Hemorrhagic Fever, Ebola/prevention & control , Viral Envelope Proteins/immunology , Adolescent , Adult , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytokines/immunology , Double-Blind Method , Ebola Vaccines/adverse effects , Ebola Vaccines/genetics , Ebolavirus/genetics , Ebolavirus/immunology , Enzyme-Linked Immunosorbent Assay , Female , Hemorrhagic Fever, Ebola/immunology , Humans , Male , Middle Aged , Neutralization Tests , Placebos/administration & dosage , T-Lymphocytes/immunology , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Envelope Proteins/genetics , Young Adult
8.
J Immunol Methods ; 362(1-2): 180-4, 2010 Oct 31.
Article in English | MEDLINE | ID: mdl-20727896

ABSTRACT

Respiratory syncytial virus (RSV) is an important cause of respiratory infection in people of all ages, and is the leading cause of hospitalization in infants. Although commercially available monoclonal antibody is available for passive prophylaxis of neonates at risk of severe disease, there is no available vaccine to prevent RSV. Measurement of neutralizing activity will be a key endpoint for vaccine evaluation. Assessment of neutralizing antibody against RSV has been limited to traditional plaque reduction, which is time-consuming and inherently operator dependent and highly variable. Here, we describe a flow cytometry-based RSV-specific neutralization assay which is more rapid than traditional methods, highly sensitive and highly reproducible.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Flow Cytometry/methods , Respiratory Syncytial Virus Infections/blood , Respiratory Syncytial Viruses , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Humans , Infant , Infant, Newborn , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/immunology , Sensitivity and Specificity
9.
Mucosal Immunol ; 3(5): 475-86, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20555315

ABSTRACT

Mucosal immunization may be important for protection against pathogens whose transmission and pathogenesis target the mucosal tissue. The capsid proteins of human papillomavirus (HPV) confer tropism for the basal epithelium and can encapsidate DNA during self-assembly to form pseudovirions (PsVs). Therefore, we produced mucosal vaccine vectors by HPV PsV encapsidation of DNA plasmids expressing an experimental antigen derived from the M and M2 proteins of respiratory syncytial virus (RSV). Intravaginal (IVag) delivery elicited local and systemic M-M2-specific CD8+ T-cell and antibody responses in mice that were comparable to an approximately 10,000-fold higher dose of naked DNA. A single HPV PsV IVag immunization primed for M-M2-specific-IgA in nasal and vaginal secretions. Based on light emission and immunofluorescent microscopy, immunization with HPV PsV-encapsidated luciferase- and red fluorescent protein (RFP)-expressing plasmids resulted in transient antigen expression (<5 days), which was restricted to the vaginal epithelium. HPV PsV encapsidation of plasmid DNA is a novel strategy for mucosal immunization that could provide new vaccine options for selected mucosal pathogens.


Subject(s)
Mucous Membrane/metabolism , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Viruses/physiology , Virion/metabolism , Administration, Intravaginal , Administration, Mucosal , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Epithelium/immunology , Epithelium/metabolism , Epithelium/pathology , Epithelium/virology , Female , Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Lymphocyte Activation , Mice , Mice, Inbred Strains , Mucous Membrane/immunology , Mucous Membrane/pathology , Mucous Membrane/virology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/transmission , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/pathogenicity , Vaccines, DNA , Vagina/pathology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Viral Matrix Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , Virion/genetics , Virion/pathogenicity
10.
Acta Virol ; 53(3): 207-11, 2009.
Article in English | MEDLINE | ID: mdl-19953728

ABSTRACT

Human respiratory syncytial virus (HRSV) associated with bronchiolitis and asthma is known to replicate actively in ciliated epithelial cells. However, little is known about the influence of HRSV replication on the cell cycle. We found that HRSV infection of HEp-2 cells led to a reduction of the number of cells in S-phase, to an increase in the number of cells in G1-phase, together with an increase of GADD153 mRNA levels and GADD153 protein expression. These results implied that a shorter S-phase supported HRSV replication suggesting possible strategies for interfering with productive HRSV infection.


Subject(s)
RNA, Messenger/analysis , Respiratory Syncytial Virus, Human/physiology , S Phase , Transcription Factor CHOP/genetics , Cell Line , G1 Phase , Humans , Virus Replication
13.
Thorax ; 57(6): 524-7, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12037228

ABSTRACT

BACKGROUND: Smooth muscle contraction is one of the hallmarks of asthma. A recently developed pyridine derivative, Y-27632, a selective Rho kinase inhibitor, has been reported to inhibit the smooth muscle contraction of human and animal trachea in ex vivo systems but its effect in animal models of airway hyperresponsiveness (AHR) has not been examined. The purpose of this study was to evaluate the effect of Y-27632 in a murine model of allergic and virally induced AHR. METHODS: Baseline lung resistance and methacholine induced AHR were measured in mice sensitised to ovalbumin (OVA) and also in mice infected with respiratory syncytial virus (RSV) following ovalbumin sensitisation (OVA/RSV). RESULTS: Time course and dose ranging experiments indicated that 30 mg/kg Y-27632 given by gavage 2 hours before methacholine challenge significantly reduced baseline lung resistance and prevented AHR in OVA sensitised mice. Y-27632 also suppressed AHR induced by the bronchospastic agent serotonin in OVA sensitised mice and prevented methacholine induced AHR in OVA/RSV mice. CONCLUSIONS: These results suggest that the signalling pathway mediated through Rho kinase may have an important role in bronchial smooth muscle tone in allergen induced and virus induced AHR and should be considered as a novel target for asthma treatment.


Subject(s)
Antihypertensive Agents/therapeutic use , Benzopyrans/therapeutic use , Bronchial Hyperreactivity/drug therapy , Respiratory Syncytial Virus Infections/complications , Airway Resistance/drug effects , Animals , Asthma/drug therapy , Asthma/physiopathology , Bronchial Hyperreactivity/etiology , Dose-Response Relationship, Drug , Female , Lung/drug effects , Mice , Mice, Inbred BALB C , Ovalbumin
14.
J Infect Dis ; 184(11): 1374-9, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11709778

ABSTRACT

Severe respiratory syncytial virus (RSV) infection has been hypothesized to be a risk factor for the development of allergy and asthma, but epidemiologic studies in humans have been inconclusive. By use of a well-characterized murine model of RSV infection and allergic sensitization with ovalbumin, the effect of a preceding severe RSV infection on the development of the pulmonary allergic inflammatory response and airway hyperresponsiveness (AHR) was tested. The impact of prior allergic sensitization on RSV-induced illness, as measured by weight loss, also was evaluated. RSV infection before allergic sensitization decreased allergen-induced AHR, production of interleukin-13 in lung tissue, and lung eosinophilia. In contrast, allergic sensitization before RSV infection increased AHR and decreased RSV-related weight loss and lung levels of interferon-gamma but did not alter viral clearance. These data provide evidence that RSV-associated AHR occurs in hosts with allergic responses and that allergic inflammation is diminished when preceded by RSV infection.


Subject(s)
Hypersensitivity/immunology , Ovalbumin/immunology , Respiratory Syncytial Virus Infections/complications , Allergens/immunology , Animals , Bronchial Hyperreactivity/diagnosis , Bronchial Hyperreactivity/virology , Bronchoconstrictor Agents , Female , Hypersensitivity/complications , Interferon-gamma/biosynthesis , Interleukin-13/biosynthesis , Lung/immunology , Lung/virology , Lymphocyte Count , Methacholine Chloride , Mice , Mice, Inbred BALB C , Pulmonary Eosinophilia/complications , Pulmonary Eosinophilia/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/growth & development , Time Factors , Virus Replication , Weight Loss
16.
Respir Res ; 2(4): 245-9, 2001.
Article in English | MEDLINE | ID: mdl-11686890

ABSTRACT

The interaction between viruses and dendritic cells (DCs) is varied and complex. DCs are key elements in the development of a host response to pathogens such as viruses, but viruses have developed survival tactics to either evade or diminish the immune system that functions to kill and eliminate these micro-organisms. In the present review we summarize current concepts regarding the function of DCs in the immune system, our understanding of how viruses alter DC function to attenuate both the virus-specific and global immune response, and how we may be able to exploit DC function to prevent or treat viral infections.


Subject(s)
Dendritic Cells/physiology , Lung/immunology , Virus Diseases/immunology , Animals , Antigen-Presenting Cells/physiology , Humans
17.
J Virol ; 75(20): 9918-24, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11559824

ABSTRACT

Virus-specific cytotoxic T lymphocytes are key effectors for the clearance of virus-infected cells and are required for the normal clearance of respiratory syncytial virus (RSV) in mice. Although perforin/granzyme-mediated lysis of infected cells is thought to be the major molecular mechanism used by CD8(+) cytotoxic T lymphocytes for elimination of virus, its role in RSV has not been reported. Here, we show that viral clearance in perforin knockout (PKO) mice is slightly delayed but that both PKO and wild-type mice clear virus by day 10, suggesting an alternative mechanism of RSV clearance. Effector T cells from the lungs of both groups of mice were shown to lyse Fas (CD95)-overexpressing target cells in greater numbers than target cells expressing low levels of Fas, suggesting that Fas ligand (CD95L)-mediated target cell lysis was occurring in vivo. This cell lysis was associated with a delay in RSV-induced disease in PKO mice compared to the time of disease onset for wild-type controls, which correlated with increased and prolonged production of gamma interferon and tumor necrosis factor alpha levels in PKO mice. We conclude that while perforin is not necessary for the clearance of primary RSV infection, the use of alternative CTL target cell killing mechanisms is less efficient and can lead to enhanced disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Membrane Glycoproteins/deficiency , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses , Animals , Disease Models, Animal , Female , Interferon-gamma/analysis , Lung/immunology , Lung/virology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Perforin , Pore Forming Cytotoxic Proteins , Respiratory Syncytial Virus Infections/virology , Tumor Necrosis Factor-alpha/analysis , fas Receptor/analysis
18.
Virology ; 283(2): 188-96, 2001 May 10.
Article in English | MEDLINE | ID: mdl-11336544

ABSTRACT

Respiratory syncytial virus (RSV) is an important human pathogen that can cause severe and life-threatening respiratory infections in infants and immunocompromised adults. We have recently shown the RSV F glycoprotein, which mediates viral fusion and entry, interacts with the cellular protein RhoA in two-hybrid and in vitro binding assays. Whether this interaction occurs in living cells remains an open question. However, because RhoA signaling is associated with many cellular functions relevant to RSV pathogenesis such as actin cytoskeleton organization, expression of proinflammatory cytokines, and smooth muscle contraction, we asked whether RhoA activation occurred during RSV infection of HEp-2 cells. We found that the amount of isoprenylated and membrane-bound RhoA in RSV-infected cultures was increased. Further evidence of RhoA activation was demonstrated by downstream signaling activity mediated by RhoA. There was an increase in p130(cas) phosphorylation during RSV infection, which was prevented by Y-27632, a specific inhibitor of Rho kinase, or lovastatin, an HMG-CoA reductase inhibitor that reduces the synthesis of groups needed for isoprenylation. In addition, RSV infection of HEp-2 cells resulted in an increase in the formation of actin stress fibers. Pretreatment of HEp-2 cells with Clostridium botulinum C3 exotoxin, an enzyme that specifically ADP-ribosylates and inactivates RhoA, prevented RSV-induced stress fiber formation. These observations indicate that RhoA and subsequent downstream signaling events are activated during RSV infection, which has implications for RSV pathogenesis.


Subject(s)
Gene Expression Regulation , Proteins , Respiratory Syncytial Virus Infections/physiopathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Cell Membrane/metabolism , Crk-Associated Substrate Protein , Fluorescent Antibody Technique , Humans , Phosphoproteins/metabolism , Phosphorylation , Protein Prenylation , Retinoblastoma-Like Protein p130 , Tumor Cells, Cultured , Virus Replication , rhoA GTP-Binding Protein/genetics
19.
Vaccine ; 19(15-16): 2080-91, 2001 Feb 28.
Article in English | MEDLINE | ID: mdl-11228380

ABSTRACT

Three separate studies were undertaken in HIV-1 uninfected persons to determine if the adjuvant QS-21 improves the magnitude or kinetics of immune responses induced by recombinant soluble gp120 HIV-1(MN) protein (rsgp120) immunization. The QS-21 was administered at two doses (50 and 100 microg), either alone or in combination with aluminum hydroxide (600 microg). At the highest doses of rsgp120 (100, 300, and 600 microg), QS-21 exerted no significant effect on either binding or neutralizing antibody titers. Antibody binding and neutralizing responses fell dramatically when rsgp120, formulated with alum alone, was given at low doses (3 and 30 microg). In contrast, antibody responses similar in titer to those in the high dose antigen groups were induced with the low dose rsgp120 formulated with QS-21. In addition, the lymphocyte proliferation and delayed type hypersensitivity skin testing were superior in the QS-21 recipients compared with the alum recipients at the low antigen doses. Moderate to severe pain was observed in majority of the volunteers receiving QS-21 formulations, and vasovagal episodes and hypertension were not infrequent. Thus, the use of QS-21 may provide a means to reduce the dose of a soluble protein immunogen.


Subject(s)
AIDS Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , HIV Envelope Protein gp120/administration & dosage , Saponins/administration & dosage , AIDS Vaccines/adverse effects , AIDS Vaccines/isolation & purification , Adolescent , Adult , Aluminum Hydroxide/administration & dosage , Animals , CHO Cells , Cricetinae , HIV Antibodies/biosynthesis , HIV Envelope Protein gp120/isolation & purification , Humans , Hypersensitivity, Delayed , Immunization , In Vitro Techniques , Lymphocyte Activation , Middle Aged , Safety , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/isolation & purification
20.
Cutis ; 67(2): 117-20, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11236220

ABSTRACT

Choriocarcinoma, a malignancy of trophoblastic cells, is characterized by the secretion of human chorionic gonadotropin (hCG). Choriocarcinoma primarily arises from the fetal (placental) trophoblasts in the setting of a molar pregnancy. Nongestational choriocarcinoma from the ovary or testis is much rarer. Testicular choriocarcinoma is a malignant tumor with great propensity for distant metastasis. The primary sites of metastasis are the lungs, liver, and brain. Skin metastasis is very rare but portends a grave prognosis when diagnosed. We present the case of a 24-year-old white male with a testicular mixed germ-cell tumor with skin metastases of choriocarcinoma.


Subject(s)
Choriocarcinoma/secondary , Skin Neoplasms/secondary , Testicular Neoplasms/pathology , Adult , Fatal Outcome , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...