Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 188
Filter
1.
Front Immunol ; 12: 724763, 2021.
Article in English | MEDLINE | ID: mdl-34489978

ABSTRACT

Characterizing the serologic features of asymptomatic SARS-CoV-2 infection is imperative to improve diagnostics and control of SARS-CoV-2 transmission. In this study, we evaluated the antibody profiles in 272 plasma samples collected from 59 COVID-19 patients, consisting of 18 asymptomatic patients, 33 mildly ill patients and 8 severely ill patients. We measured the IgG against five viral structural proteins, different isotypes of immunoglobulins against the Receptor Binding Domain (RBD) protein, and neutralizing antibodies. The results showed that the overall antibody response was lower in asymptomatic infections than in symptomatic infections throughout the disease course. In contrast to symptomatic patients, asymptomatic patients showed a dominant IgG-response towards the RBD protein, but not IgM and IgA. Neutralizing antibody titers had linear correlations with IgA/IgM/IgG levels against SARS-CoV-2-RBD, as well as with IgG levels against multiple SARS-CoV-2 structural proteins, especially with anti-RBD or anti-S2 IgG. In addition, the sensitivity of anti-S2-IgG is better in identifying asymptomatic infections at early time post infection compared to anti-RBD-IgG. These data suggest that asymptomatic infections elicit weaker antibody responses, and primarily induce IgG antibody responses rather than IgA or IgM antibody responses. Detection of IgG against the S2 protein could supplement nucleic acid testing to identify asymptomatic patients. This study provides an antibody detection scheme for asymptomatic infections, which may contribute to epidemic prevention and control.


Subject(s)
Antibodies, Viral/blood , Asymptomatic Infections , Immunoglobulin G/blood , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Viral Structural Proteins/immunology , Adolescent , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/physiology , Binding Sites, Antibody , Female , Humans , Immunoglobulin G/classification , Immunoglobulin M/immunology , Kinetics , Male , Middle Aged , Neutralization Tests/statistics & numerical data , SARS-CoV-2/chemistry , Young Adult
2.
JCI Insight ; 6(6)2021 03 22.
Article in English | MEDLINE | ID: mdl-33621211

ABSTRACT

Comorbid medical illnesses, such as obesity and diabetes, are associated with more severe COVID-19, hospitalization, and death. However, the role of the immune system in mediating these clinical outcomes has not been determined. We used multiparameter flow cytometry and systems serology to comprehensively profile the functions of T cells and antibodies targeting spike, nucleocapsid, and envelope proteins in a convalescent cohort of COVID-19 subjects who were either hospitalized (n = 20) or not hospitalized (n = 40). To avoid confounding, subjects were matched by age, sex, ethnicity, and date of symptom onset. Surprisingly, we found that the magnitude and functional breadth of virus-specific CD4+ T cell and antibody responses were consistently higher among hospitalized subjects, particularly those with medical comorbidities. However, an integrated analysis identified more coordination between polyfunctional CD4+ T cells and antibodies targeting the S1 domain of spike among subjects who were not hospitalized. These data reveal a functionally diverse and coordinated response between T cells and antibodies targeting SARS-CoV-2, which is reduced in the presence of comorbid illnesses that are known risk factors for severe COVID-19.


Subject(s)
Antibodies, Viral/physiology , CD4-Positive T-Lymphocytes/physiology , COVID-19/virology , Hospitalization , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus , Virion , Adult , Aged , Antibodies, Neutralizing/metabolism , Antibodies, Neutralizing/physiology , Antibodies, Viral/metabolism , CD4-Positive T-Lymphocytes/metabolism , COVID-19/epidemiology , COVID-19/immunology , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/immunology , Comorbidity , Diabetes Mellitus/epidemiology , Diabetes Mellitus/immunology , Female , Humans , Immunity, Humoral , Male , Middle Aged , Nucleocapsid , Severity of Illness Index , Viral Envelope , Viral Proteins , Young Adult
3.
J Med Virol ; 93(5): 3158-3164, 2021 May.
Article in English | MEDLINE | ID: mdl-33590900

ABSTRACT

Data regarding the immunological memory and long-time kinetics of immunoglobulin (IgG) against viral nucleoprotein (NP) and spike protein S1 receptor-binding domain (S1RBD) of Severe Acute Respiratory Syndrome-associated Coronavirus 2 (SARS-CoV-2) are lacking. All consecutive COVID-19 patients admitted to our Clinic between March 1, 2020, and May 1, 2020, who were tested at hospital admission for anti-S1RBD and anti-NP IgG were enrolled. Serum samples were tested for anti-SARS-CoV-2 antibodies with the use of two commercially available enzyme-linked immunosorbent assays. Results are expressed as optical density measurements at 450 nm (OD450 ). Overall, 111 patients were included; the median (q1-q3) age was 57 (49-73) years, 59 (53%) males. According to disease severity, 31 (28%), 47 (42%), and 33 (30%) patients were considered affected by mild/moderate, severe, and critical SARS-CoV-2 infection, respectively. During hospitalization, patients with the critical disease showed a higher peak value of both anti-NP (median OD450 : 3.66 vs. 3.06 vs. 3.00 respectively, p = .043) and anti-S1RBD IgG (median OD450 : 2.33 vs. 1.6 vs. 0.91, respectively, p < .001). By testing 48 subjects 6 months or above from discharge, a significant decrease of anti-NP IgG was observed (r: -0.5838; p < .0001), whereas anti-S1RBD IgG showed only a modest reduction (r: -0.1507; p = .0647). Accordingly, 10 (21%) and 2 (4%) patients had a negative serological status for anti-NP and anti-S1RBD IgG, respectively; no association with clinical severity was found. IgGs against SARS-CoV-2 persisted several months after discharge, regardless of disease severity, suggesting that vaccination could be a valid strategy to fight the pandemic.


Subject(s)
Antibodies, Viral/physiology , COVID-19/immunology , COVID-19/pathology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Aged , Female , Humans , Male , Middle Aged , Protein Binding , Protein Domains , SARS-CoV-2/metabolism
4.
Poult Sci ; 100(2): 603-614, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33518113

ABSTRACT

Pigeon paramyxovirus type 1 (PPMV-1) is a globally distributed, virulent member of the avian paramyxovirus type-1. The PPMV-1-associated disease poses a great threat to the pigeon industry. The innate immune response is crucial for antiviral infections and revealing the pathogenic mechanisms of PPMV-1. In this study, we evaluated the pathogenicity of a PPMV-1 strain LHLJ/110822 in one-month-old domestic pigeons, as well as the host immune responses in PPMV-1-infected pigeons. We observed typically clinical sign in infected pigeons by 3 dpi. The morbidity rate and the mortality in pigeons inoculated with the PPMV-1 strain were up to 100% and 30%, respectively. The virus could replicate in all of the examined tissues, namely trachea, lung, liver, spleen, and bursa of Fabricius. In addition, the infected pigeons had developed anti-PPMV-1 antibodies as early as 8 dpi; and the antibody level increased over the time in this study. The expression level of toll-like receptor (TLR) 2, TLR3 TLR15, IFN-γ, and IL-6 were significantly upregulated by the PPMV-1 infection in some tissues of pigeons. By contrast, PPMV-1 infection results in downregulation of IL-18 expression in most of investigated tissues except for bursa of Fabricius in this study. The current results confirmed that this virus could replicate in pigeons and induce host immune responses, then leading to produce serum antibody titers. Meanwhile, the PPMV-1 infection induces strong innate immune responses and intense inflammatory responses at early stage in pigeon which may associate with the viral pathogenesis.


Subject(s)
Columbidae , Newcastle Disease/immunology , Newcastle disease virus/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Viral/physiology , Chick Embryo , Eggs/virology , Immunity, Innate , Newcastle disease virus/pathogenicity , Specific Pathogen-Free Organisms
7.
J Vis Exp ; (158)2020 04 29.
Article in English | MEDLINE | ID: mdl-32420984

ABSTRACT

Commensal bacteria are well established to impact infection of eukaryotic viruses. Direct binding between the pathogen and the host microbiome is responsible for altering infection for many of these viruses. Thus, characterizing the nature of virus-bacteria binding is a foundational step needed for elucidating the mechanism(s) by which bacteria alter viral infection. For human norovirus, commensal bacteria enhance B cell infection. The virus directly binds to these bacteria, indicating that this direct interaction is involved in the mechanism of infection enhancement. A variety of techniques can be used to quantify interactions between bacteria and viruses including scintillation counting of radiolabeled viruses and polymerase chain reaction (PCR). Both methods require the use of live virus, which may need to be generated in the laboratory. Currently, none of the established in vitro culture systems available for human norovirus are robust enough to allow for generation of highly concentrated viral stocks. In lieu of live virus, virus-like particles (VLPs) have been used to characterize the interactions between norovirus and bacteria. Herein a flow cytometry method is described with uses virus specific antibodies to quantify VLP binding to gram-negative and gram-positive bacteria. Inclusion of both bacteria only and isotype controls allowed for optimization of the assay to reduce background antibody binding and accurate quantification of VLP attachment to the bacteria tested. High VLP:bacterium ratios result in VLPs binding to large percentages of the bacterial population. However, when VLP quantities are decreased, the percent of bacteria bound also decreases. Ultimately, this method can be employed in future experiments elucidating the specific conditions and structural components that regulate norovirus:bacterial interactions.


Subject(s)
Gram-Negative Bacteria/physiology , Gram-Positive Bacteria/physiology , Microbial Interactions , Norovirus/physiology , Virus Attachment , Antibodies, Viral/physiology , Flow Cytometry
8.
Am J Trop Med Hyg ; 101(1): 207-213, 2019 07.
Article in English | MEDLINE | ID: mdl-31162004

ABSTRACT

rVSVΔG-ZEBOV-GP vaccine is a live recombinant (r) vesicular stomatitis virus (VSV), where the VSV G protein is replaced with the Zaire Ebola virus (ZEBOV) glycoprotein (GP). For vaccine immunogenicity testing, clinical trial sera collected during an active ZEBOV outbreak underwent gamma irradiation (GI) before testing in biosafety level 2 laboratories to inactivate possible wild-type ZEBOV. Before irradiating pivotal trial samples, two independent studies evaluated the impact of GI (50 kGy) on binding ZEBOV-GP (ELISA) antibodies against rVSVΔG-ZEBOV-GP, using sera from a North American phase 1 study. Gamma irradiation was associated with slightly higher antibody concentrations in pre-vaccination samples and slightly lower concentrations postvaccination. Results indicate that GI is a viable method for treating samples from regions where filoviruses are endemic, with minor effects on antibody titers. The impact of GI on immunogenicity analyses should be considered when interpreting data from irradiated specimens.


Subject(s)
Antibodies, Viral/radiation effects , Ebola Vaccines/immunology , Ebolavirus/metabolism , Gamma Rays , Serum/radiation effects , Antibodies, Viral/blood , Antibodies, Viral/physiology , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Humans , Membrane Glycoproteins , Vaccination , Vaccines, Synthetic/immunology , Viral Envelope Proteins/immunology
9.
Am J Trop Med Hyg ; 101(1): 198-206, 2019 07.
Article in English | MEDLINE | ID: mdl-31074409

ABSTRACT

Filoviruses, which include ebolaviruses and marburgvirus, can cause outbreaks of highly lethal hemorrhagic fever. This disease causes significant morbidity and mortality in humans and non-human primates, with human fatality rates reaching 90% during some outbreaks. Currently, there is lack of licensed vaccines or antivirals for these viruses. Since early symptoms of filovirus infection mimic more common diseases, there is a strong unmet public health and biodefense need for broad-spectrum filovirus rapid diagnostics. We have generated a panel of mouse single-chain Fv-antibodies (scFvs) to filovirus glycoproteins (GPs) using cell-free ribosome display and determined their cross-reactivity profiles to all known filovirus species. Two scFvs (4-2 and 22-1) were able to detect all known Ebolavirus and Marburgvirus species. This is the first report on ribosome display scFvs that can detect a broad set of filovirus GPs, which demonstrates the potential for use in diagnostics.


Subject(s)
Antibodies, Viral/physiology , Filoviridae/immunology , Animals , Antibody Affinity , Antibody Specificity , Cell-Free System , Cloning, Molecular , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Ribosomes , Single-Chain Antibodies , Viral Proteins/immunology
10.
Clin Infect Dis ; 69(1): 151-154, 2019 06 18.
Article in English | MEDLINE | ID: mdl-30561563

ABSTRACT

Human immunodeficiency virus (HIV)-exposed, uninfected infants have higher risks of respiratory syncytial virus-associated hospitalization than HIV-unexposed infants. Despite similar neutralizing antibody titers between HIV-infected and -uninfected women, maternal HIV infection and hypergammaglobulinemia were independently associated with lower titers in newborns. Maternal hypergammaglobulinemia was associated with lower cord-to-maternal antibody ratio.


Subject(s)
Antibodies, Neutralizing/physiology , Antibodies, Viral/physiology , HIV Infections/complications , Immunity, Maternally-Acquired , Respiratory Syncytial Virus, Human/immunology , Female , Humans , Hypergammaglobulinemia , Infant, Newborn , Pregnancy
11.
PLoS Biol ; 16(8): e2006601, 2018 08.
Article in English | MEDLINE | ID: mdl-30096134

ABSTRACT

Determining the duration of protective immunity requires quantifying the magnitude and rate of loss of antibodies to different virus and vaccine antigens. A key complication is heterogeneity in both the magnitude and decay rate of responses of different individuals to a given vaccine, as well as of a given individual to different vaccines. We analyzed longitudinal data on antibody titers in 45 individuals to characterize the extent of this heterogeneity and used models to determine how it affected the longevity of protective immunity to measles, rubella, vaccinia, tetanus, and diphtheria. Our analysis showed that the magnitude of responses in different individuals varied between 12- and 200-fold (95% coverage) depending on the antigen. Heterogeneity in the magnitude and decay rate contribute comparably to variation in the longevity of protective immunity between different individuals. We found that some individuals have, on average, slightly longer-lasting memory than others-on average, they have higher antibody levels with slower decay rates. We identified different patterns for the loss of protective levels of antibodies to different vaccine and virus antigens. Specifically, we found that for the first 25 to 50 years, virtually all individuals have protective antibody titers against diphtheria and tetanus, respectively, but about 10% of the population subsequently lose protective immunity per decade. In contrast, at the outset, not all individuals had protective titers against measles, rubella, and vaccinia. However, these antibody titers wane much more slowly, with a loss of protective immunity in only 1% to 3% of the population per decade. Our results highlight the importance of long-term longitudinal studies for estimating the duration of protective immunity and suggest both how vaccines might be improved and how boosting schedules might be reevaluated.


Subject(s)
Antibodies, Viral/physiology , Antibodies/physiology , Immunologic Memory/physiology , Adolescent , Adult , Antibodies/metabolism , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Immunization, Secondary , Immunologic Memory/immunology , Longitudinal Studies , Male , Viruses/immunology , Young Adult
12.
PLoS Negl Trop Dis ; 12(8): e0006685, 2018 08.
Article in English | MEDLINE | ID: mdl-30092029

ABSTRACT

BACKGROUND: Dengue is the most prevalent arthropod-borne viral human disease in tropical and subtropical regions, caused by four dengue virus (DENV) serotypes. In spite of the increasing global incidence, no specific antiviral therapy is available. Cells of the mononuclear phagocyte lineage are the main targets either for direct antibody (Ab)-independent or Ab-mediated human DENV infection, usually associated to the severe forms of disease. Since the virus entry may be a convenient therapeutic alternative, this study aimed to investigate the mode of DENV internalization into myeloid cells in the absence and presence of DENV Ab and evaluate the inhibitory activity of diverse biochemical inhibitors of endocytosis. METHODOLOGY/PRINCIPAL FINDINGS: By infectivity assays and quantitative RT-PCR determinations, it was demonstrated that DENV-2 entry into U937 and K562 cells in the absence of Ab was highly inhibited by the early treatment with ammonium chloride, chlorpromazine and dynasore, but it was not affected by methyl-ß-cyclodextrin, indicating that DENV-2 utilizes a low pH-dependent, clathrin- and dynamin-mediated endocytic infectious pathway for the direct entry into both human myeloid cells. To study the Ab-mediated entry of DENV, the experimental conditions for enhancement of infection were established by inoculating immune complexes formed with DENV-2 and the Ab 2H2 or 3H5. The internalization of DENV-2-2H2 or DENV-2-3H5 complexes in both myeloid cells was also dependent on acid pH and dynamin but a differential requirement of the clathrin-mediated endocytic route was observed depending on the FcγR involved in the complex uptake: the infection through FcγRII was dependent on clathrin-coated vesicles whereas the internalization pathway mediated by FcγRI was independent of clathrin. This property was not serotype-specific. CONCLUSIONS/SIGNIFICANCE: DENV entry into myeloid cells in the absence or presence of Ab can be blocked by diverse biochemical inhibitors affecting the cellular factors involved in endocytosis. The identification of the virus-host interactions involved in virus penetration may allow the finding of host-targeted antivirals widely active against diverse pathogenic flaviviruses with similar requirements for virus entry.


Subject(s)
Antibodies, Viral/physiology , Dengue Virus/physiology , Endocytosis/drug effects , Myeloid Cells/virology , Virus Internalization/drug effects , Ammonium Chloride , Cell Survival , Chlorpromazine/pharmacology , Humans , Hydrazones/pharmacology , K562 Cells , Myeloid Cells/drug effects , U937 Cells , beta-Cyclodextrins/pharmacology
13.
Epidemiol Infect ; 146(11): 1372-1383, 2018 08.
Article in English | MEDLINE | ID: mdl-29909804

ABSTRACT

Previously we reported on the HPIV2 genotype distribution in Croatia 2011-2014. Here we expand this period up to 2017 and confirm that G1a genotype has replaced G3 genotype from the period 2011-2014. Our hypothesis was that the G1a-to-G3 genotype replacement is an antibody-driven event. A cross-neutralisation with anti-HPIV2 sera specific for either G1a or G3 genotype revealed the presence of genotype-specific antigenic determinants. By the profound, in silico analyses three potential B cell epitopic regions were identified in the hemagglutinin neuraminidase (regions 314-361 and 474-490) and fusion protein (region 440-484). The region identified in the fusion protein does not show any unique site between the G1a and G3 isolates, five differentially glycosylated sites in the G1a and G3 genotype isolates were identified in epitopic regions of hemagglutinin neuraminidase. All positively selected codons were found to be located either in the region 314-316 or in the region 474-490 what indicates a strong positive selection in this region and reveals that these regions are susceptible to evolutionary pressure possibly caused by antibodies what gives a strong verification to our hypothesis that neutralising antibodies are a key determinant in the inherently complex adaptive evolution of HPIV2 in the region.


Subject(s)
Antibodies, Neutralizing/physiology , Parainfluenza Virus 2, Human/genetics , Rubulavirus Infections/virology , Adolescent , Age Distribution , Animals , Antibodies, Viral/physiology , Child , Child, Preschool , Chlorocebus aethiops , Croatia/epidemiology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Female , Genotype , Guinea Pigs , HN Protein/immunology , Humans , Infant , Likelihood Functions , Middle Aged , Parainfluenza Virus 2, Human/classification , Parainfluenza Virus 2, Human/immunology , Phylogeny , RNA, Viral/chemistry , RNA, Viral/genetics , Recurrence , Rubulavirus Infections/epidemiology , Rubulavirus Infections/immunology , Seasons , Sequence Alignment , Vero Cells
14.
Avian Pathol ; 46(4): 434-441, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28290220

ABSTRACT

Despite the widespread and successful use of Newcastle disease (ND) vaccines, Newcastle disease virus (NDV) can seriously injure the reproductive tract of egg-laying hens, leading to rapid egg-drop and poor shell quality. Few published studies investigated local NDV-specific immune response in the reproductive tract after ND vaccination of hens. The present study investigated, for the first time, local NDV-specific antibody-mediated immunity in segments of the oviduct during the laying period. Specific pathogen-free (SPF) White Leghorn chickens were immunized following an ND vaccination programme applied in the field, which combined ND-attenuated vaccine (inoculated subcutaneously at one day, 2 weeks and 11 weeks of age) with inactivated vaccine (inoculated intramuscularly at 17 weeks). The infundibulum, magnum, isthmus and uterus (segments of the reproductive tract) were harvested at 28 weeks and 32 weeks of age (during the laying period). Supernatant from ex vivo tissue culture was collected and tested by: (i) haemagglutination inhibition (HI) test, (ii) commercial IDVet ND-enzyme-linked immunosorbent assay (ELISA) and (iii) NDV-specific IgG, IgM and IgA in-house ELISAs. For all sampling time points and oviduct segments, all samples were positive for commercial ND-ELISA and in-house ELISA-IgG. However, six of these ELISA-IgG positive samples yielded negative results when submitted to the HI test. Interestingly, NDV-specific IgM and IgA were detected frequently in the infundibulum and magnum as compared to the isthmus and uterus. These results show that the antibody immune response in the oviduct was induced by the timing of attenuated and inactivated ND vaccinations.


Subject(s)
Antibodies, Viral/physiology , Chickens , Genitalia, Female/immunology , Newcastle Disease/prevention & control , Newcastle disease virus/immunology , Viral Vaccines/immunology , Animals , Antibody Specificity , Female , Oviposition , Poultry Diseases/prevention & control , Poultry Diseases/virology , Specific Pathogen-Free Organisms , Vaccines, Attenuated/immunology , Vaccines, Inactivated/immunology
15.
Acta Virol ; 60(3): 271-80, 2016.
Article in English | MEDLINE | ID: mdl-27640437

ABSTRACT

Porcine circovirus type 2 (PCV2) is the primary viral pathogen of porcine circovirus associated disease (PCVAD) and vaccination is an important method to prevent and control the disease. The expression of PCV2 capsid protein (Cap) in adenovirus vector system has been investigated, but the poor immune responses limit its application. In this study, transcriptional enhancer element largest intron of the human cytomegalovirus (Intron A) and woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) were applied to increase the immunogenicity of PCV2 Cap adenovirus-based vaccine. Western blot and indirect immunofluorescence assay (IFA) analysis showed that modified adenoviruses with Intron A and WPRE alone or both could significantly increase the expression of Cap compared to the unmodified adenoviruses. Furthermore, the humoral and cellular immune responses of the constructed recombinant adenoviruses were evaluated in mice. Indirect ELISA, virus neutralizing test and western blot showed that modified adenoviruses elicited higher humoral immune responses than unmodified adenovirus, and Intron A-WPRE-modified virus immunized group had better immune response than the others. Besides, the results of lymphocyte proliferation response and cytokines release assay showed that enhanced cellular immune responses were induced by modified adenoviruses. These results demonstrated that Intron A and WPRE significantly improved the expression of the Cap protein in adenovirus vector system and enhanced the immune responses in mice, making the adenovirus vector system more applicable against PCV2.


Subject(s)
Adenoviridae/genetics , Antibodies, Viral/physiology , Circovirus/metabolism , Animals , Cell Line , Cell Proliferation , Circoviridae Infections/prevention & control , Circoviridae Infections/virology , Circovirus/classification , Circovirus/genetics , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation, Viral/physiology , HEK293 Cells , Humans , Lymphocytes/physiology , Lymphocytes/virology , Mice , Swine , Viral Vaccines/immunology
16.
Nat Commun ; 7: 10844, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26936020

ABSTRACT

The Fc region of HIV-1 Env-specific broadly neutralizing antibodies (bNAbs) is required for suppressing viraemia, through mechanisms which remain poorly understood. Here, we identify bNAbs that exert antibody-dependent cellular cytotoxicity (ADCC) in cell culture and kill HIV-1-infected lymphocytes through natural killer (NK) engagement. These antibodies target the CD4-binding site, the glycans/V3 and V1/V2 loops on gp120, or the gp41 moiety. The landscape of Env epitope exposure at the surface and the sensitivity of infected cells to ADCC vary considerably between viral strains. Efficient ADCC requires sustained cell surface binding of bNAbs to Env, and combining bNAbs allows a potent killing activity. Furthermore, reactivated infected cells from HIV-positive individuals expose heterogeneous Env epitope patterns, with levels that are often but not always sufficient to trigger killing by bNAbs. Our study delineates the parameters controlling ADCC activity of bNAbs, and supports the use of the most potent antibodies to clear the viral reservoir.


Subject(s)
Antibodies, Neutralizing/physiology , Antibodies, Viral/physiology , CD4-Positive T-Lymphocytes/physiology , HIV-1/physiology , Animals , Cell Line , Humans
17.
J Clin Invest ; 125(3): 1255-68, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25689254

ABSTRACT

The emergence and seasonal persistence of pathogenic H7N9 influenza viruses in China have raised concerns about the pandemic potential of this strain, which, if realized, would have a substantial effect on global health and economies. H7N9 viruses are able to bind to human sialic acid receptors and are also able to develop resistance to neuraminidase inhibitors without a loss in fitness. It is not clear whether prior exposure to circulating human influenza viruses or influenza vaccination confers immunity to H7N9 strains. Here, we demonstrate that 3 of 83 H3 HA-reactive monoclonal antibodies generated by individuals that had previously undergone influenza A virus vaccination were able to neutralize H7N9 viruses and protect mice against homologous challenge. The H7N9-neutralizing antibodies bound to the HA stalk domain but exhibited a difference in their breadth of reactivity to different H7 influenza subtypes. Mapping viral escape mutations suggested that these antibodies bind at least two different epitopes on the stalk region. Together, these results indicate that these broadly neutralizing antibodies may contribute to the development of therapies against H7N9 strains and may also be effective against pathogenic H7 strains that emerge in the future.


Subject(s)
Antibodies, Neutralizing/physiology , Antibodies, Viral/physiology , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/prevention & control , Vaccination , Animals , Antibodies, Monoclonal/physiology , Cross Reactions , Dogs , Female , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H7N9 Subtype/genetics , Influenza Vaccines , Influenza, Human/immunology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Neutralization Tests , Point Mutation
18.
Gene Ther ; 22(2): 116-26, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25503696

ABSTRACT

Gene delivery vectors based on adeno-associated viruses (AAV) have exhibited promise in both preclinical disease models and human clinical trials for numerous disease targets, including the retinal degenerative disorders Leber's congenital amaurosis and choroideremia. One general challenge for AAV is that preexisting immunity, as well as subsequent development of immunity following vector administration, can severely inhibit systemic AAV vector gene delivery. However, the role of neutralizing antibodies (NABs) in AAV transduction of tissues considered to be immune privileged, such as the eye, is unclear in large animals. Intravitreal AAV administration allows for broad retinal delivery, but is more susceptible to interactions with the immune system than subretinal administration. To assess the effects of systemic anti-AAV antibody levels on intravitreal gene delivery, we quantified the anti-AAV antibodies present in sera from non-human primates before and after intravitreal injections with various AAV capsids. Analysis showed that intravitreal administration resulted in an increase in anti-AAV antibodies regardless of the capsid serotype, transgene or dosage of virus injected. For monkeys injected with wild-type AAV2 and/or an AAV2 mutant, the variable that most significantly affected the production of anti-AAV2 antibodies was the amount of virus delivered. In addition, post-injection antibody titers were highest against the serotype administered, but the antibodies were also cross-reactive against other AAV serotypes. Furthermore, NAB levels in serum correlated with those in vitreal fluid, demonstrating both that this route of administration exposes AAV capsid epitopes to the adaptive immune system and that serum measurements are predictive of vitreous fluid NAB titers. Moreover, the presence of preexisting NAB titers in the serum of monkeys correlated strongly (R=0.76) with weak, decaying or no transgene expression following intravitreal administration of AAV. Investigating anti-AAV antibody development will aid in understanding the interactions between gene therapy vectors and the immune system during ocular administration and can form a basis for future clinical studies applying intravitreal gene delivery.


Subject(s)
Antibodies, Neutralizing/physiology , Antibodies, Viral/physiology , Dependovirus/immunology , Retinal Degeneration/therapy , Animals , Dependovirus/genetics , Genetic Therapy , Genetic Vectors , HEK293 Cells , Humans , Intravitreal Injections , Macaca mulatta , Transduction, Genetic
19.
PLoS One ; 9(7): e102415, 2014.
Article in English | MEDLINE | ID: mdl-25019613

ABSTRACT

The Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) is the etiological agent for the infectious disease, SARS, which first emerged 10 years ago. SARS-CoV is a zoonotic virus that has crossed the species barriers to infect humans. Bats, which harbour a diverse pool of SARS-like CoVs (SL-CoVs), are believed to be the natural reservoir. The SARS-CoV surface Spike (S) protein is a major antigenic determinant in eliciting neutralizing antibody production during SARS-CoV infection. In our previous work, we showed that a panel of murine monoclonal antibodies (mAbs) that target the S2 subunit of the S protein are capable of neutralizing SARS-CoV infection in vitro (Lip KM et al, J Virol. 2006 Jan; 80(2): 941-50). In this study, we report our findings on the characterization of one of these mAbs, known as 1A9, which binds to the S protein at a novel epitope within the S2 subunit at amino acids 1111-1130. MAb 1A9 is a broadly neutralizing mAb that prevents viral entry mediated by the S proteins of human and civet SARS-CoVs as well as bat SL-CoVs. By generating mutant SARS-CoV that escapes the neutralization by mAb 1A9, the residue D1128 in S was found to be crucial for its interaction with mAb 1A9. S protein containing the substitution of D1128 with alanine (D1128A) exhibited a significant decrease in binding capability to mAb 1A9 compared to wild-type S protein. By using a pseudotyped viral entry assay, it was shown that the D1128A substitution in the escape virus allows it to overcome the viral entry blockage by mAb 1A9. In addition, the D1128A mutation was found to exert no effects on the S protein cell surface expression and incorporation into virion particles, suggesting that the escape virus retains the same viral entry property as the wild-type virus.


Subject(s)
Amino Acid Substitution , Antibodies, Neutralizing/physiology , Aspartic Acid/chemistry , Severe acute respiratory syndrome-related coronavirus/genetics , Spike Glycoprotein, Coronavirus/genetics , Amino Acid Sequence , Animals , Antibodies, Monoclonal/physiology , Antibodies, Viral/physiology , CHO Cells , Chiroptera/virology , Chlorocebus aethiops , Cricetulus , Epitope Mapping , HEK293 Cells , Humans , Mice, Inbred BALB C , Molecular Sequence Data , Neutralization Tests , Severe acute respiratory syndrome-related coronavirus/metabolism , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Sequence Alignment , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Vero Cells , Virion/genetics , Virion/pathogenicity , Viverridae/virology
20.
PLoS One ; 9(6): e99905, 2014.
Article in English | MEDLINE | ID: mdl-24937158

ABSTRACT

Newcastle disease virus (NDV) is a member of the Paramyxovirinae subfamily and can infect most species of birds. It has been a great threat for the poultry industry all around the world. In this report, we successfully produced infectious pseudotyped pNL4-3-Luc-R(-)E(-) (HIV-Luc) viruses with the HN and F envelope proteins of NDV. Further investigation revealed the cytoplasmic domains of HN and F, especially HN, plays a significant role in the infection efficiency of these pseudotyped HIV-Luc viruses. Replacement of, or direct fusion to the cytoplasmic domain of the HN protein by that of vesicular stomatitis virus G (VSV-G) could greatly enhance or destroy the infective potential of HN and F-pseudotyped (NDV-pseudotyped) HIV-Luc virus. We further established a novel neutralization assay to evaluate neutralizing antibodies against NDV with the NDV-pseudotyped HIV-Luc viruses. Comparative neutralization data indicate that the results determined by using the NDV-pseudotyped HIV-Luc viruses are as reliable as those by the conventional virus-neutralization assay (VN test) with native NDV. Moreover, the results show that the novel neutralization assay is more sensitive than the VN test.


Subject(s)
HIV-1/genetics , Newcastle Disease/immunology , Newcastle disease virus/immunology , Animals , Antibodies, Neutralizing/physiology , Antibodies, Viral/physiology , Birds , Genes, Reporter , HEK293 Cells , HN Protein/genetics , Humans , Immune Sera/immunology , Luciferases/biosynthesis , Luciferases/genetics , Neutralization Tests , Newcastle Disease/prevention & control , Newcastle Disease/virology , Newcastle disease virus/genetics , Viral Fusion Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...