64 results on '"Sangster MY"'
Search Results
2. Association of Human Milk Antibody Induction, Persistence, and Neutralizing Capacity With SARS-CoV-2 Infection vs mRNA Vaccination.
- Author
-
Young BE, Seppo AE, Diaz N, Rosen-Carole C, Nowak-Wegrzyn A, Cruz Vasquez JM, Ferri-Huerta R, Nguyen-Contant P, Fitzgerald T, Sangster MY, Topham DJ, and Järvinen KM
- Subjects
- Adult, Cohort Studies, Female, Humans, Immunoglobulin A immunology, Immunoglobulin G immunology, Infant, Lactation, Male, Antibodies, Neutralizing blood, COVID-19 immunology, COVID-19 Vaccines immunology, Milk, Human immunology, SARS-CoV-2 immunology
- Abstract
Importance: Long-term effect of parental COVID-19 infection vs vaccination on human milk antibody composition and functional activity remains unclear., Objective: To compare temporal IgA and IgG response in human milk and microneutralization activity against SARS-CoV-2 between lactating parents with infection and vaccinated lactating parents out to 90 days after infection or vaccination., Design, Setting, and Participants: Convenience sampling observational cohort (recruited July to December 2020) of lactating parents with infection with human milk samples collected at days 0 (within 14 days of diagnosis), 3, 7, 10, 28, and 90. The observational cohort included vaccinated lactating parents with human milk collected prevaccination, 18 days after the first dose, and 18 and 90 days after the second dose., Exposures: COVID-19 infection diagnosed by polymerase chain reaction within 14 days of consent or receipt of messenger RNA (mRNA) COVID-19 vaccine (BNT162b2 or mRNA-1273)., Main Outcomes and Measures: Human milk anti-SARS-CoV-2 receptor-binding domain IgA and IgG and microneutralization activity against live SARS-CoV-2 virus., Results: Of 77 individuals, 47 (61.0%) were in the infection group (mean [SD] age, 29.9 [4.4] years), and 30 (39.0%) were in the vaccinated group (mean [SD] age, 33.0 [3.4] years; P = .002). The mean (SD) age of infants in the infection and vaccinated group were 3.1 (2.2) months and 7.5 (5.2) months, respectively (P < .001). Infection was associated with a variable human milk IgA and IgG receptor-binding domain-specific antibody response over time that was classified into different temporal patterns: upward trend and level trend (33 of 45 participants [73%]) and low/no response (12 of 45 participants [27%]). Infection was associated with a robust and quick IgA response in human milk that was stable out to 90 days after diagnosis. Vaccination was associated with a more uniform IgG-dominant response with concentrations increasing after each vaccine dose and beginning to decline by 90 days after the second dose. Vaccination was associated with increased human milk IgA after the first dose only (mean [SD] increase, 31.5 [32.6] antibody units). Human milk collected after infection and vaccination exhibited microneutralization activity. Microneutralization activity increased throughout time in the vaccine group only (median [IQR], 2.2 [0] before vaccine vs 10 [4.0] after the first dose; P = .003) but was higher in the infection group (median [IQR], 20 [67] at day 28) vs the vaccination group after the first-dose human milk samples (P = .002). Both IgA and non-IgA (IgG-containing) fractions of human milk from both participants with infection and those who were vaccinated exhibited microneutralization activity against SARS-CoV-2., Conclusions and Relevance: In this cohort study of a convenience sample of lactating parents, the pattern of IgA and IgG antibodies in human milk differed between COVID-19 infection vs mRNA vaccination out to 90 days. While infection was associated with a highly variable IgA-dominant response and vaccination was associated with an IgG-dominant response, both were associated with having human milk that exhibited neutralization activity against live SARS-CoV-2 virus.
- Published
- 2022
- Full Text
- View/download PDF
3. Formation and Expansion of Memory B Cells against Coronavirus in Acutely Infected COVID-19 Individuals.
- Author
-
Embong AK, Nguyen-Contant P, Wang J, Kanagaiah P, Chaves FA, Fitzgerald TF, Zhou Q, Kosoy G, Branche AR, Miller BL, Zand MS, Sangster MY, and Topham DJ
- Abstract
Infection with the β-coronavirus SARS-CoV-2 typically generates strong virus-specific antibody production. Antibody responses against novel features of SARS-CoV-2 proteins require naïve B cell activation, but there is a growing appreciation that conserved regions are recognized by pre-existing memory B cells (MBCs) generated by endemic coronaviruses. The current study investigated the role of pre-existing cross-reactive coronavirus memory in the antibody response to the viral spike (S) and nucleocapsid (N) proteins following SARS-CoV-2 infection. The breadth of reactivity of circulating antibodies, plasmablasts, and MBCs was analyzed. Acutely infected subjects generated strong IgG responses to the S protein, including the novel receptor binding domain, the conserved S2 region, and to the N protein. The response included reactivity to the S of endemic β-coronaviruses and, interestingly, to the N of an endemic α-coronavirus. Both mild and severe infection expanded IgG MBC populations reactive to the S of SARS-CoV-2 and endemic β-coronaviruses. Avidity of S-reactive IgG antibodies and MBCs increased after infection. Overall, findings indicate that the response to the S and N of SARS-CoV-2 involves pre-existing MBC activation and adaptation to novel features of the proteins, along with the potential of imprinting to shape the response to SARS-CoV-2 infection.
- Published
- 2022
- Full Text
- View/download PDF
4. Broadly Reactive IgG Responses to Heterologous H5 Prime-Boost Influenza Vaccination Are Shaped by Antigenic Relatedness to Priming Strains.
- Author
-
Wang J, Li D, Perry S, Hilchey SP, Wiltse A, Treanor JJ, Sangster MY, and Zand MS
- Subjects
- Adult, Antibodies, Viral immunology, Cohort Studies, Hemagglutinin Glycoproteins, Influenza Virus immunology, Humans, Immunoglobulin G immunology, Influenza A Virus, H5N1 Subtype genetics, Influenza Vaccines administration & dosage, Middle Aged, Antibodies, Viral blood, Antigenic Drift and Shift, Immunoglobulin G blood, Influenza A Virus, H5N1 Subtype immunology, Influenza Vaccines immunology, Influenza, Human prevention & control, Vaccination methods
- Abstract
Prime-boost vaccinations of humans with different H5 strains have generated broadly protective antibody levels. However, the effect of an individual's H5 exposure history on antibody responses to subsequent H5 vaccination is poorly understood. To investigate this, we analyzed the IgG responses to H5 influenza A/Indonesia/5/2005 (Ind05) virus vaccination in three cohorts: (i) a doubly primed group that had received two H5 virus vaccinations, namely, against influenza A/Vietnam/203/2004 (Vie04) virus 5 years prior and A/Hong Kong/156/1997 (HK97) 11 years prior to the Ind05 vaccination; (ii) a singly primed group that had received a vaccination against Vie04 virus 5 years prior to the Ind05 vaccination; and (iii) an H5-naive group that received two doses of the Ind05 vaccine 28 days apart. Hemagglutinin (HA)-reactive IgG levels were estimated by a multiplex assay against an HA panel that included 21 H5 strains and 9 other strains representing the H1, H3, H7, and H9 subtypes. Relative HA antibody landscapes were generated to quantitatively analyze the magnitude and breadth of antibody binding after vaccination. We found that short-interval priming and boosting with the Ind05 vaccine in the naive group generated a low anti-H5 response. Both primed groups generated robust antibody responses reactive to a broad range of H5 strains after receiving a booster injection of Ind05 vaccine; IgG antibody levels persisted longer in subjects who had been doubly primed years ago. Notably, the IgG responses were strongest against the first priming H5 strain, which reflects influenza virus immune imprinting. Finally, the broad anti-H5 IgG response was stronger against strains having a small antigenic distance from the initial priming strain. IMPORTANCE The antigenic shift and draft of hemagglutinin (HA) in influenza viruses is accepted as one of the major reasons for immune evasion. The analysis of B cell immune responses to influenza infection and vaccination is complicated by the impact of exposure history and antibody cross-reactions between antigenically similar influenza strains. To assist in such analyses, the influenza "antibody landscape" method has been used to analyze and visualize the relationship of antibody-mediated immunity to antigenic distances between influenza strains. In this study, we describe a "relative antibody landscape" method that calculates the antigenic distance between the vaccine influenza strain and other H5 strains and uses this relative antigenic distance to plot the anti-H5 IgG levels postvaccination. This new method quantitatively estimates and visualizes the correlation between the humoral response to a particular influenza strain and the antigenic distance from other strains. Our findings demonstrate the effect of a subject's H5 exposure history on H5 vaccine responses quantified by the relative antibody landscape method.
- Published
- 2021
- Full Text
- View/download PDF
5. Short term results of vaccination with adjuvanted recombinant varicella zoster glycoprotein E during initial BTK inhibitor therapy for CLL or lymphoplasmacytic lymphoma.
- Author
-
Zent CS, Brady MT, Delage C, Strawderman M, Laniewski N, Contant PN, Kanagaiah P, Sangster MY, Barr PM, Chu CC, Topham DJ, and Friedberg JW
- Subjects
- Adjuvants, Immunologic administration & dosage, Follow-Up Studies, Humans, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Middle Aged, Prognosis, Prospective Studies, Waldenstrom Macroglobulinemia immunology, Waldenstrom Macroglobulinemia pathology, Agammaglobulinaemia Tyrosine Kinase antagonists & inhibitors, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Protein Kinase Inhibitors therapeutic use, Recombinant Proteins immunology, Vaccination methods, Viral Envelope Proteins immunology, Waldenstrom Macroglobulinemia drug therapy
- Published
- 2021
- Full Text
- View/download PDF
6. Squalene-Based Influenza Vaccine Adjuvants and Their Impact on the Hemagglutinin-Specific B Cell Response.
- Author
-
Nguyen-Contant P, Sangster MY, and Topham DJ
- Abstract
Influenza infections continue to cause significant annual morbidity and mortality despite ongoing influenza vaccine research. Adjuvants are administered in conjunction with influenza vaccines to enhance the immune response and strengthen protection against disease. Squalene-based emulsion adjuvants including MF59, AS03, and AF03, are registered for administration with influenza vaccines and are widely used in many countries. Squalene-based emulsion adjuvants induce a strong innate immune response, enhancing antigen presentation both quantitively and qualitatively to generate strong B cell responses and antibody production. They also diversify the reactivity profiles and strengthen the affinities of antibodies against the influenza hemagglutinin, increasing protection across virus clades. In this review, we consider the mechanisms of the enhancement of innate and adaptive immune responses by squalene-based emulsionSE adjuvants and the resulting increase in magnitude and breadth of hemagglutinin-specific B cell responses. We relate observed effects of SE adjuvants and current mechanistic understandings to events in responding lymph nodes. These insights will guide the rational design and optimization of influenza vaccines to provide broad and effective protection.
- Published
- 2021
- Full Text
- View/download PDF
7. Immunity to Influenza Infection in Humans.
- Author
-
Topham DJ, DeDiego ML, Nogales A, Sangster MY, and Sant A
- Subjects
- Age Factors, Animals, CD8-Positive T-Lymphocytes immunology, Disease Models, Animal, Humans, Influenza, Human immunology, Mice, Adaptive Immunity, Antibodies, Viral immunology, Influenza Vaccines immunology, Influenza, Human prevention & control
- Abstract
This review discusses the human immune responses to influenza infection with some insights from studies using animal models, such as experimental infection of mice. Recent technological advances in the study of human immune responses have greatly added to our knowledge of the infection and immune responses, and therefore much of the focus is on recent studies that have moved the field forward. We consider the complexity of the adaptive response generated by many sequential encounters through infection and vaccination., (Copyright © 2021 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
8. Characterization of SARS-CoV-2 RNA, Antibodies, and Neutralizing Capacity in Milk Produced by Women with COVID-19.
- Author
-
Pace RM, Williams JE, Järvinen KM, Belfort MB, Pace CDW, Lackey KA, Gogel AC, Nguyen-Contant P, Kanagaiah P, Fitzgerald T, Ferri R, Young B, Rosen-Carole C, Diaz N, Meehan CL, Caffé B, Sangster MY, Topham D, McGuire MA, Seppo A, and McGuire MK
- Subjects
- Adult, Breast virology, Breast Feeding, COVID-19 transmission, COVID-19 virology, Female, Humans, Infant, Infectious Disease Transmission, Vertical, Milk, Human virology, Mothers, Pregnancy, Pregnancy Complications, Infectious virology, RNA, Viral isolation & purification, SARS-CoV-2 isolation & purification, Antibodies, Neutralizing metabolism, Antibodies, Viral metabolism, COVID-19 immunology, Milk, Human immunology, Pregnancy Complications, Infectious immunology, SARS-CoV-2 immunology
- Abstract
Whether mother-to-infant SARS-CoV-2 transmission can occur during breastfeeding and, if so, whether the benefits of breastfeeding outweigh this risk during maternal COVID-19 illness remain important questions. Using RT-qPCR, we did not detect SARS-CoV-2 RNA in any milk sample ( n = 37) collected from 18 women following COVID-19 diagnosis. Although we detected evidence of viral RNA on 8 out of 70 breast skin swabs, only one was considered a conclusive positive result. In contrast, 76% of the milk samples collected from women with COVID-19 contained SARS-CoV-2-specific IgA, and 80% had SARS-CoV-2-specific IgG. In addition, 62% of the milk samples were able to neutralize SARS-CoV-2 infectivity in vitro , whereas milk samples collected prior to the COVID-19 pandemic were unable to do so. Taken together, our data do not support mother-to-infant transmission of SARS-CoV-2 via milk. Importantly, milk produced by infected mothers is a beneficial source of anti-SARS-CoV-2 IgA and IgG and neutralizes SARS-CoV-2 activity. These results support recommendations to continue breastfeeding during mild-to-moderate maternal COVID-19 illness. IMPORTANCE Results from prior studies assaying human milk for the presence of SARS-CoV-2, the causative virus of COVID-19, have suggested milk may act as a potential vehicle for mother-to-child transmission. Most previous studies are limited because they followed only a few participants, were cross-sectional, and/or failed to report how milk was collected and/or analyzed. As such, considerable uncertainty remains regarding whether human milk is capable of transmitting SARS-CoV-2 from mother to child. Here, we report that repeated milk samples collected from 18 women following COVID-19 diagnosis did not contain SARS-CoV-2 RNA; however, risk of transmission via breast skin should be further evaluated. Importantly, we found that milk produced by infected mothers is a source of anti-SARS-CoV-2 IgA and IgG and neutralizes SARS-CoV-2 activity. These results support recommendations to continue breastfeeding during mild-to-moderate maternal COVID-19 illness as milk likely provides specific immunologic benefits to infants., (Copyright © 2021 Pace et al.)
- Published
- 2021
- Full Text
- View/download PDF
9. Array-based analysis of SARS-CoV-2, other coronaviruses, and influenza antibodies in convalescent COVID-19 patients.
- Author
-
Steiner DJ, Cognetti JS, Luta EP, Klose AM, Bucukovski J, Bryan MR, Schmuke JJ, Nguyen-Contant P, Sangster MY, Topham DJ, and Miller BL
- Subjects
- Betacoronavirus isolation & purification, Biosensing Techniques instrumentation, Biosensing Techniques methods, COVID-19, Coronavirus Infections diagnosis, Coronavirus Infections virology, Equipment Design, HEK293 Cells, Humans, Influenza, Human diagnosis, Influenza, Human virology, Middle East Respiratory Syndrome Coronavirus isolation & purification, Pandemics, Pneumonia, Viral diagnosis, Pneumonia, Viral virology, Protein Array Analysis instrumentation, Protein Array Analysis methods, Severe acute respiratory syndrome-related coronavirus isolation & purification, SARS-CoV-2, Sensitivity and Specificity, Antibodies, Viral blood, Coronavirus isolation & purification, Coronavirus Infections blood, Influenza A virus isolation & purification, Influenza, Human blood, Pneumonia, Viral blood
- Abstract
Detection of antibodies to upper respiratory pathogens is critical to surveillance, assessment of the immune status of individuals, vaccine development, and basic biology. The urgent need for antibody detection tools has proven particularly acute in the COVID-19 era. We report a multiplex label-free antigen microarray on the Arrayed Imaging Reflectometry (AIR) platform for detection of antibodies to SARS-CoV-2, SARS-CoV-1, MERS, three circulating coronavirus strains (HKU1, 229E, OC43) and three strains of influenza. We find that the array is readily able to distinguish uninfected from convalescent COVID-19 subjects, and provides quantitative information about total Ig, as well as IgG- and IgM-specific responses., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
10. Analysis of Antigen-Specific Human Memory B Cell Populations Based on In Vitro Polyclonal Stimulation.
- Author
-
Nguyen-Contant P, Embong AK, Topham DJ, and Sangster MY
- Subjects
- Animals, Epitopes, Humans, Immunization, Immunologic Memory, Mice, B-Lymphocyte Subsets immunology, B-Lymphocytes immunology, Cell Culture Techniques, Enzyme-Linked Immunospot Assay methods
- Abstract
Antigen-specific memory B cell (MBC) populations mediate the rapid, strong, and high-affinity secondary antibody responses that play a key role in combating infection and generating protective responses to vaccination. Recently, cell staining with fluorochrome-labeled antigens together with sequencing methods such as Drop-seq and CITE-seq have provided information on the specificity, phenotype, and transcriptome of single MBCs. However, characterization of MBCs at the level of antigen-reactive populations remains an important tool for assessing an individual's B cell immunity and responses to antigen exposure. This is readily performed using a long-established method based on in vitro polyclonal stimulation of MBCs to induce division and differentiation into antibody-secreting cells (ASCs). Post-stimulation antigen-specific measurement of the MBC-derived ASCs (or the secreted antibodies) indicates the size of precursor MBC populations. Additional information about the character of antigen-reactive MBC populations is provided by analysis of MBC-derived antibodies of particular specificities for binding avidity and functionality. This article outlines a simple and reliable strategy for efficient in vitro MBC stimulation and use of the ELISpot assay as a post-stimulation readout to determine the size of antigen-specific MBC populations. Other applications of the in vitro stimulation technique for MBC analysis are discussed. The following protocols are included. © 2020 Wiley Periodicals LLC Basic Protocol 1: Polyclonal stimulation of memory B cells using unfractionated PBMCs Alternate Protocol: Stimulation of small PBMC numbers using 96-well plates with U-bottom wells Basic Protocol 2: ELISpot assay for enumeration of memory B cell-derived antibody-secreting cells., (© 2020 Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
11. S Protein-Reactive IgG and Memory B Cell Production after Human SARS-CoV-2 Infection Includes Broad Reactivity to the S2 Subunit.
- Author
-
Nguyen-Contant P, Embong AK, Kanagaiah P, Chaves FA, Yang H, Branche AR, Topham DJ, and Sangster MY
- Subjects
- Adult, Antibodies, Viral immunology, B-Lymphocytes virology, COVID-19, Convalescence, Coronavirus Nucleocapsid Proteins, Cross Reactions, Female, Healthy Volunteers, Humans, Immunologic Memory, Male, Middle Aged, Nucleocapsid Proteins immunology, Pandemics, Phosphoproteins, Protein Interaction Domains and Motifs, Protein Subunits, SARS-CoV-2, Spike Glycoprotein, Coronavirus chemistry, B-Lymphocytes immunology, Betacoronavirus immunology, Coronavirus Infections immunology, Immunoglobulin G immunology, Pneumonia, Viral immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
The high susceptibility of humans to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the cause of coronavirus disease 2019 (COVID-19), reflects the novelty of the virus and limited preexisting B cell immunity. IgG against the SARS-CoV-2 spike (S) protein, which carries the novel receptor binding domain (RBD), is absent or at low levels in unexposed individuals. To better understand the B cell response to SARS-CoV-2 infection, we asked whether virus-reactive memory B cells (MBCs) were present in unexposed subjects and whether MBC generation accompanied virus-specific IgG production in infected subjects. We analyzed sera and peripheral blood mononuclear cells (PBMCs) from non-SARS-CoV-2-exposed healthy donors and COVID-19 convalescent subjects. Serum IgG levels specific for SARS-CoV-2 proteins (S, including the RBD and S2 subunit, and nucleocapsid [N]) and non-SARS-CoV-2 proteins were related to measurements of circulating IgG MBC levels. Anti-RBD IgG was absent in unexposed subjects. Most unexposed subjects had anti-S2 IgG, and a minority had anti-N IgG, but IgG MBCs with these specificities were not detected, perhaps reflecting low frequencies. Convalescent subjects had high levels of IgG against the RBD, S2, and N, together with large populations of RBD- and S2-reactive IgG MBCs. Notably, IgG titers against the S protein of the human coronavirus OC43 were higher in convalescent subjects than in unexposed subjects and correlated strongly with anti-S2 titers. Our findings indicate cross-reactive B cell responses against the S2 subunit that might enhance broad coronavirus protection. Importantly, our demonstration of MBC induction by SARS-CoV-2 infection suggests that a durable form of B cell immunity is maintained even if circulating antibody levels wane. IMPORTANCE The recent rapid worldwide spread of SARS-CoV-2 has established a pandemic of potentially serious disease in the highly susceptible human population. Key issues are whether humans have preexisting immune memory that provides some protection against SARS-CoV-2 and whether SARS-CoV-2 infection generates lasting immune protection against reinfection. Our analysis focused on pre- and postinfection IgG and IgG memory B cells (MBCs) reactive to SARS-CoV-2 proteins. Most importantly, we demonstrate that infection generates both IgG and IgG MBCs against the novel receptor binding domain and the conserved S2 subunit of the SARS-CoV-2 spike protein. Thus, even if antibody levels wane, long-lived MBCs remain to mediate rapid antibody production. Our study results also suggest that SARS-CoV-2 infection strengthens preexisting broad coronavirus protection through S2-reactive antibody and MBC formation., (Copyright © 2020 Nguyen-Contant et al.)
- Published
- 2020
- Full Text
- View/download PDF
12. COVID-19 and human milk: SARS-CoV-2, antibodies, and neutralizing capacity.
- Author
-
Pace RM, Williams JE, Järvinen KM, Belfort MB, Pace CDW, Lackey KA, Gogel AC, Nguyen-Contant P, Kanagaiah P, Fitzgerald T, Ferri R, Young B, Rosen-Carole C, Diaz N, Meehan CL, Caffe B, Sangster MY, Topham D, McGuire MA, Seppo A, and McGuire MK
- Abstract
Background: It is not known whether SARS-CoV-2 can be transmitted from mother to infant during breastfeeding, and if so whether the benefits of breastfeeding outweigh this risk. This study was designed to evaluate 1) if SARS-CoV-2 RNA can be detected in milk and on the breast of infected women, 2) concentrations of milk-borne anti-SARS-CoV-2 antibodies, and 3) the capacity of milk to neutralize SARS-CoV-2 infectivity., Methods: We collected 37 milk samples and 70 breast swabs (before and after breast washing) from 18 women recently diagnosed with COVID-19. Samples were analyzed for SARS-CoV-2 RNA using RT-qPCR. Milk was also analyzed for IgA and IgG specific for the nucleocapsid protein, receptor binding domain (RBD), S2 subunit of the spike protein of SARS-CoV-2, as well as 2 seasonal coronaviruses using ELISA; and for its ability to neutralize SARS-CoV-2., Results: We did not detect SARS-CoV-2 RNA in any milk sample. In contrast, SARS-CoV-2 RNA was detected on several breast swabs, although only one was considered conclusive. All milk contained SARS-CoV-2-specific IgA and IgG, and levels of anti-RBD IgA correlated with SARS-CoV-2 neutralization. Strong correlations between levels of IgA and IgG to SARS-CoV-2 and seasonal coronaviruses were noted., Conclusions: Our data do not support maternal-to-child transmission of SARS-CoV-2 via milk; however, risk of transmission via breast skin should be further evaluated. Importantly, milk produced by infected mothers is a source of anti-SARS-CoV-2 IgA and IgG and neutralizes SARS-CoV-2 activity. These results support recommendations to continue breastfeeding during mild-to-moderate maternal COVID-19 illness.
- Published
- 2020
- Full Text
- View/download PDF
13. Implementing sequence-based antigenic distance calculation into immunological shape space model.
- Author
-
Anderson CS, Sangster MY, Yang H, Mariani TJ, Chaudhury S, and Topham DJ
- Subjects
- Amino Acid Sequence, Antibodies, Viral blood, Antigens, Viral chemistry, Antigens, Viral immunology, Computer Simulation, Cross Reactions, Hemagglutinin Glycoproteins, Influenza Virus chemistry, Humans, Antibodies, Viral immunology, Hemagglutinin Glycoproteins, Influenza Virus immunology, Influenza A Virus, H1N1 Subtype immunology, Influenza Vaccines immunology, Models, Immunological
- Abstract
Background: In 2009, a novel influenza vaccine was distributed worldwide to combat the H1N1 influenza "swine flu" pandemic. However, antibodies induced by the vaccine display differences in their specificity and cross-reactivity dependent on pre-existing immunity. Here, we present a computational model that can capture the effect of pre-existing immunity on influenza vaccine responses. The model predicts the region of the virus hemagglutinin (HA) protein targeted by antibodies after vaccination as well as the level of cross-reactivity induced by the vaccine. We tested our model by simulating a scenario similar to the 2009 pandemic vaccine and compared the results to antibody binding data obtained from human subjects vaccinated with the monovalent 2009 H1N1 influenza vaccine., Results: We found that both specificity and cross-reactivity of the antibodies induced by the 2009 H1N1 influenza HA protein were affected by the viral strain the individual was originally exposed. Specifically, the level of antigenic relatedness between the original exposure HA antigen and the 2009 HA protein affected antigenic-site immunodominance. Moreover, antibody cross-reactivity was increased when the individual's pre-existing immunity was specific to an HA protein antigenically distinct from the 2009 pandemic strain. Comparison of simulation data with antibody binding data from human serum samples demonstrated qualitative and quantitative similarities between the model and real-life immune responses to the 2009 vaccine., Conclusion: We provide a novel method to evaluate expected outcomes in antibody specificity and cross-reactivity after influenza vaccination in individuals with different influenza HA antigen exposure histories. The model produced similar outcomes as what has been previously reported in humans after receiving the 2009 influenza pandemic vaccine. Our results suggest that differences in cross-reactivity after influenza vaccination should be expected in individuals with different exposure histories.
- Published
- 2020
- Full Text
- View/download PDF
14. Characterizing Emerging Canine H3 Influenza Viruses.
- Author
-
Martinez-Sobrido L, Blanco-Lobo P, Rodriguez L, Fitzgerald T, Zhang H, Nguyen P, Anderson CS, Holden-Wiltse J, Bandyopadhyay S, Nogales A, DeDiego ML, Wasik BR, Miller BL, Henry C, Wilson PC, Sangster MY, Treanor JJ, Topham DJ, Byrd-Leotis L, Steinhauer DA, Cummings RD, Luczo JM, Tompkins SM, Sakamoto K, Jones CA, Steel J, Lowen AC, Danzy S, Tao H, Fink AL, Klein SL, Wohlgemuth N, Fenstermacher KJ, El Najjar F, Pekosz A, Sauer L, Lewis MK, Shaw-Saliba K, Rothman RE, Liu ZY, Chen KF, Parrish CR, Voorhees IEH, Kawaoka Y, Neumann G, Chiba S, Fan S, Hatta M, Kong H, Zhong G, Wang G, Uccellini MB, García-Sastre A, Perez DR, Ferreri LM, Herfst S, Richard M, Fouchier R, Burke D, Pattinson D, Smith DJ, Meliopoulos V, Freiden P, Livingston B, Sharp B, Cherry S, Dib JC, Yang G, Russell CJ, Barman S, Webby RJ, Krauss S, Danner A, Woodard K, Peiris M, Perera RAPM, Chan MCW, Govorkova EA, Marathe BM, Pascua PNQ, Smith G, Li YT, Thomas PG, and Schultz-Cherry S
- Subjects
- Animals, Communicable Diseases, Emerging transmission, Communicable Diseases, Emerging virology, Dog Diseases transmission, Dogs, Ferrets, Guinea Pigs, Humans, Influenza A Virus, H3N2 Subtype classification, Influenza A Virus, H3N2 Subtype genetics, Influenza A Virus, H3N8 Subtype classification, Influenza A Virus, H3N8 Subtype genetics, Influenza A virus classification, Influenza A virus genetics, Influenza, Human transmission, Influenza, Human virology, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Inbred DBA, United States, Zoonoses transmission, Communicable Diseases, Emerging veterinary, Dog Diseases virology, Influenza A Virus, H3N2 Subtype isolation & purification, Influenza A Virus, H3N8 Subtype isolation & purification, Influenza A virus isolation & purification, Zoonoses virology
- Abstract
The continual emergence of novel influenza A strains from non-human hosts requires constant vigilance and the need for ongoing research to identify strains that may pose a human public health risk. Since 1999, canine H3 influenza A viruses (CIVs) have caused many thousands or millions of respiratory infections in dogs in the United States. While no human infections with CIVs have been reported to date, these viruses could pose a zoonotic risk. In these studies, the National Institutes of Allergy and Infectious Diseases (NIAID) Centers of Excellence for Influenza Research and Surveillance (CEIRS) network collaboratively demonstrated that CIVs replicated in some primary human cells and transmitted effectively in mammalian models. While people born after 1970 had little or no pre-existing humoral immunity against CIVs, the viruses were sensitive to existing antivirals and we identified a panel of H3 cross-reactive human monoclonal antibodies (hmAbs) that could have prophylactic and/or therapeutic value. Our data predict these CIVs posed a low risk to humans. Importantly, we showed that the CEIRS network could work together to provide basic research information important for characterizing emerging influenza viruses, although there were valuable lessons learned., Competing Interests: AG-S. is inventor of patents on influenza virus vaccines owned by the Icahn School for Medicine at Mount Sinai and licensed to Medimmune, BI Vetmedica, Vivaldi Biosciences, Zoetis and Avimex.
- Published
- 2020
- Full Text
- View/download PDF
15. Role of Memory B Cells in Hemagglutinin-Specific Antibody Production Following Human Influenza A Virus Infection.
- Author
-
Sangster MY, Nguyen PQT, and Topham DJ
- Abstract
When influenza A virus infects an immune individual, preexisting memory B cell (MBC) activation and rapid anamnestic antibody production plays a key role in viral clearance. The most effective neutralizing antibodies target the antigenically variable head of the viral hemagglutinin (HA); antibodies against the conserved HA stalk provide broader but less potent protection. In this review, we provide a comprehensive picture of an adult's HA-specific antibody response to influenza virus infection. The process is followed from preexisting HA-specific MBC activation and rapid production of anti-HA antibodies, through to germinal center seeding and adaptation of the response to novel features of the HA. A major focus of the review is the role of competition between preexisting MBCs in determining the character of the HA-reactive antibody response. HA novelty modifies this competition and can shift the response from the immunodominant head to the stalk. We suggest that antibodies resulting from preexisting MBC activation are important regulators of anti-HA antibody production and play a role in positive selection of germinal center B cells reactive to novel HA epitopes. Our review also considers the role of MBCs in the effects of early-life imprinting on HA head- and stalk-specific antibody responses to influenza infection. An understanding of the processes described in this review will guide development of vaccination strategies that provide broadly effective protection.
- Published
- 2019
- Full Text
- View/download PDF
16. Broad Hemagglutinin-Specific Memory B Cell Expansion by Seasonal Influenza Virus Infection Reflects Early-Life Imprinting and Adaptation to the Infecting Virus.
- Author
-
Tesini BL, Kanagaiah P, Wang J, Hahn M, Halliley JL, Chaves FA, Nguyen PQT, Nogales A, DeDiego ML, Anderson CS, Ellebedy AH, Strohmeier S, Krammer F, Yang H, Bandyopadhyay S, Ahmed R, Treanor JJ, Martinez-Sobrido L, Golding H, Khurana S, Zand MS, Topham DJ, and Sangster MY
- Subjects
- Antibodies, Viral immunology, Humans, Immunoglobulin G immunology, Influenza A Virus, H1N1 Subtype, B-Lymphocytes immunology, Hemagglutinin Glycoproteins, Influenza Virus immunology, Immunologic Memory, Influenza A Virus, H3N2 Subtype immunology, Influenza, Human immunology, Seasons
- Abstract
Memory B cells (MBCs) are key determinants of the B cell response to influenza virus infection and vaccination, but the effect of different forms of influenza antigen exposure on MBC populations has received little attention. We analyzed peripheral blood mononuclear cells and plasma collected following human H3N2 influenza infection to investigate the relationship between hemagglutinin-specific antibody production and changes in the size and character of hemagglutinin-reactive MBC populations. Infection produced increased concentrations of plasma IgG reactive to the H3 head of the infecting virus, to the conserved stalk, and to a broad chronological range of H3s consistent with original antigenic sin responses. H3-reactive IgG MBC expansion after infection included reactivity to head and stalk domains. Notably, expansion of H3 head-reactive MBC populations was particularly broad and reflected original antigenic sin patterns of IgG production. Findings also suggest that early-life H3N2 infection "imprints" for strong H3 stalk-specific MBC expansion. Despite the breadth of MBC expansion, the MBC response included an increase in affinity for the H3 head of the infecting virus. Overall, our findings indicate that H3-reactive MBC expansion following H3N2 infection is consistent with maintenance of response patterns established early in life, but nevertheless includes MBC adaptation to the infecting virus. IMPORTANCE Rapid and vigorous virus-specific antibody responses to influenza virus infection and vaccination result from activation of preexisting virus-specific memory B cells (MBCs). Understanding the effects of different forms of influenza virus exposure on MBC populations is therefore an important guide to the development of effective immunization strategies. We demonstrate that exposure to the influenza hemagglutinin via natural infection enhances broad protection through expansion of hemagglutinin-reactive MBC populations that recognize head and stalk regions of the molecule. Notably, we show that hemagglutinin-reactive MBC expansion reflects imprinting by early-life infection and that this might apply to stalk-reactive, as well as to head-reactive, MBCs. Our findings provide experimental support for the role of MBCs in maintaining imprinting effects and suggest a mechanism by which imprinting might confer heterosubtypic protection against avian influenza viruses. It will be important to compare our findings to the situation after influenza vaccination., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
17. Pandemic influenza vaccines: what they have taught us about B cell immunology.
- Author
-
Topham DJ, Nguyen P, and Sangster MY
- Subjects
- Antibodies, Viral metabolism, Cross Reactions, Hemagglutinin Glycoproteins, Influenza Virus immunology, Humans, Immunity, Humoral, Influenza A Virus, H1N1 Subtype, Influenza A Virus, H7N9 Subtype, Pandemics, B-Lymphocytes immunology, Influenza Vaccines immunology, Influenza, Human immunology, Orthomyxoviridae immunology
- Abstract
The emergence of avian influenza viruses stimulated pandemic concerns and efforts to develop protective vaccines. Studies of the immune responses to experimental vaccines for pandemic influenza have taught us lessons about human immunity to influenza in general that can be applied to seasonal, pandemic, and even universal vaccine responses. For example, the concepts of targeting the hemagglutinin stalk and elicitation of stalk reactive antibodies grew out of studies of the 2009 pandemic H1N1 vaccines. More recently, the phenomenon of imprinting, the influence of early life exposure to influenza modifying responses to the viruses or vaccines later in life, has been reinforced through the study of potential pandemic influenza virus vaccines such as H7N9. These studies have also revealed potential strategies to improve responses to novel influenza strains and produce more broadly cross-reactive B cell and antibody responses. These concepts are discussed in detail in this review., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
18. Broad cross-reactive IgG responses elicited by adjuvanted vaccination with recombinant influenza hemagglutinin (rHA) in ferrets and mice.
- Author
-
Wang J, Hilchey SP, DeDiego M, Perry S, Hyrien O, Nogales A, Garigen J, Amanat F, Huertas N, Krammer F, Martinez-Sobrido L, Topham DJ, Treanor JJ, Sangster MY, and Zand MS
- Subjects
- Animals, Cross Reactions, Ferrets, Mice, Antibodies, Viral immunology, Hemagglutinins immunology, Immunoglobulin G immunology, Influenza A Virus, H1N1 Subtype immunology, Influenza A Virus, H3N2 Subtype immunology, Influenza Vaccines, Vaccination
- Abstract
Annual immunization against influenza virus is a large international public health effort. Accumulating evidence suggests that antibody mediated cross-reactive immunity against influenza hemagglutinin (HA) strongly correlates with long-lasting cross-protection against influenza virus strains that differ from the primary infection or vaccination strain. However, the optimal strategies for achieving highly cross-reactive antibodies to the influenza virus HA have not yet to be defined. In the current study, using Luminex-based mPlex-Flu assay, developed by our laboratory, to quantitatively measure influenza specific IgG antibody mediated cross-reactivity, we found that prime-boost-boost vaccination of ferrets with rHA proteins admixed with adjuvant elicited higher magnitude and broader cross-reactive antibody responses than that induced by actual influenza viral infection, and this cross-reactive response likely correlated with increased anti-stalk reactive antibodies. We observed a similar phenomenon in mice receiving three sequential vaccinations with rHA proteins from either A/California/07/2009 (H1N1) or A/Hong Kong/1/1968 (H3N2) viruses admixed with Addavax, an MF59-like adjuvant. Using this same mouse vaccination model, we determined that Addavax plays a more significant role in the initial priming event than in subsequent boosts. We also characterized the generation of cross-reactive antibody secreting cells (ASCs) and memory B cells (MBCs) when comparing vaccination to viral infection. We have also found that adjuvant plays a critical role in the generation of long-lived ASCs and MBCs cross-reactive to influenza viruses as a result of vaccination with rHA of influenza virus, and the observed increase in stalk-reactive antibodies likely contributes to this IgG mediated broad cross-reactivity.
- Published
- 2018
- Full Text
- View/download PDF
19. Antigenicity of the 2015-2016 seasonal H1N1 human influenza virus HA and NA proteins.
- Author
-
Clark AM, DeDiego ML, Anderson CS, Wang J, Yang H, Nogales A, Martinez-Sobrido L, Zand MS, Sangster MY, and Topham DJ
- Subjects
- Animals, Dogs, Humans, Influenza, Human epidemiology, New York epidemiology, Antigens, Viral immunology, Hemagglutinin Glycoproteins, Influenza Virus immunology, Influenza A Virus, H1N1 Subtype immunology, Influenza, Human virology, Neuraminidase immunology, Seasons
- Abstract
Antigenic drift of the hemagglutinin (HA) and neuraminidase (NA) influenza virus proteins contributes to reduced vaccine efficacy. To analyze antigenic drift in human seasonal H1N1 viruses derived from the 2009 pandemic H1N1 virus (pH1N1-like viruses) accounts for the limited effectiveness (around 40%) of vaccination against pH1N1-like viruses during the 2015-2016 season, nasal washes/swabs collected from adult subjects in the Rochester, NY area, were used to sequence and isolate the circulating viruses. The HA and NA proteins from viruses circulating during the 2015-2016 season encoded eighteen and fourteen amino acid differences, respectively, when compared to A/California/04/2009, a strain circulating at the origin of the 2009 pandemic. The circulating strains belonged to subclade 6B.1, defined by HA amino acid substitutions S101N, S179N, and I233T. Hemagglutination-inhibiting (HAI) and HA-specific neutralizing serum antibody (Ab) titers from around 50% of pH1N1-like virus-infected subjects and immune ferrets were 2-4 fold lower for the 2015-2016 circulating strains compared to the vaccine strain. In addition, using a luminex-based mPlex HA assay, the binding of human sera from subjects infected with pH1N1-like viruses to the HA proteins from circulating and vaccine strains was not identical, strongly suggesting antigenic differences in the HA protein. Additionally, NA inhibition (NAI) Ab titers in human sera from pH1N1-like virus-infected subjects increased after the infection and there were measurable antigenic differences between the NA protein of circulating strains and the vaccine strain using both ferret and human antisera. Despite having been vaccinated, infected subjects exhibited low HAI Ab titers against the vaccine and circulating strains. This suggests that poor responses to the H1N1 component of the vaccine as well as antigenic differences in the HA and NA proteins of currently circulating pH1N1-like viruses could be contributing to risk of infection even after vaccination.
- Published
- 2017
- Full Text
- View/download PDF
20. Generation and Protective Ability of Influenza Virus-Specific Antibody-Dependent Cellular Cytotoxicity in Humans Elicited by Vaccination, Natural Infection, and Experimental Challenge.
- Author
-
Jegaskanda S, Luke C, Hickman HD, Sangster MY, Wieland-Alter WF, McBride JM, Yewdell JW, Wright PF, Treanor J, Rosenberger CM, and Subbarao K
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Female, Humans, Infant, Infant, Newborn, Male, Middle Aged, Young Adult, Antibodies, Viral immunology, Antibody-Dependent Cell Cytotoxicity, Influenza A Virus, H1N1 Subtype immunology, Influenza A Virus, H3N2 Subtype immunology, Influenza Vaccines immunology, Influenza, Human immunology
- Abstract
Background: Nonneutralizing antibodies (Abs) involved in antibody-dependent cellular cytotoxicity (ADCC) may provide some protection from influenza virus infection. The ability of influenza vaccines to induce ADCC-mediating Abs (ADCC-Abs) in adults and children is unclear., Methods: We quantified ADCC-Abs in serum samples from adults who received a dose of inactivated subunit vaccine (ISV) targeting monovalent 2009 pandemic influenza A(H1N1) virus or live-attenuated influenza vaccine (LAIV) or who had laboratory-confirmed influenza A(H1N1) virus infection. We also measured ADCC-Abs in children who either received a dose of trivalent seasonal ISV followed by trivalent seasonal LAIV or 2 doses of LAIV. Finally, we assessed the ability of low and high ADCC-Ab titers to protect adults from experimental challenge with influenza A/Wisconsin/67/131/2005(H3N2) virus., Results: Adults and children who received a dose of ISV had a robust increase in ADCC-Ab titers to both recombinant hemagglutinin (rHA) protein and homologous virus-infected cells. There was no detectable increase in titers of ADCC-Abs to rHA or virus-infected cells in adults and children who received LAIV. Higher titers (≥320) of preexisting ADCC-Abs were associated with lower virus replication and a significant reduction in total symptom scores in experimentally infected adults., Conclusions: ADCC-Ab titers increased following experimental influenza virus infection in adults and after ISV administration in both children and adults., (Published by Oxford University Press for the Infectious Diseases Society of America 2016. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2016
- Full Text
- View/download PDF
21. High-Affinity H7 Head and Stalk Domain-Specific Antibody Responses to an Inactivated Influenza H7N7 Vaccine After Priming With Live Attenuated Influenza Vaccine.
- Author
-
Halliley JL, Khurana S, Krammer F, Fitzgerald T, Coyle EM, Chung KY, Baker SF, Yang H, Martínez-Sobrido L, Treanor JJ, Subbarao K, Golding H, Topham DJ, and Sangster MY
- Subjects
- Antibodies, Neutralizing blood, B-Lymphocytes immunology, Cohort Studies, Hemagglutination Inhibition Tests, Humans, Influenza, Human prevention & control, Influenza, Human virology, Neutralization Tests, Vaccines, Attenuated immunology, Vaccines, Inactivated immunology, Antibodies, Viral blood, Influenza A Virus, H7N7 Subtype immunology, Influenza Vaccines immunology, Influenza, Human immunology
- Abstract
Recent studies have shown that live attenuated influenza vaccines (LAIVs) expressing avian influenza virus hemagglutinins (HAs) prime for strong protective antibody responses to an inactivated influenza vaccine (IIV) containing the HA. To better understand this priming effect, we compared H7 HA head and stalk domain-specific B-cell responses in H7N7 LAIV-primed subjects and non-H7-primed controls after a single dose of H7N7 IIV. As previously reported, H7N7 LAIV-primed subjects but not control subjects generated strong hemagglutination-inhibiting and neutralizing antibody responses to the H7N7 IIV. Here, we found that the quantity, epitope diversity, and affinity of H7 head-specific antibodies increased rapidly in only H7N7 LAIV-primed subjects after receipt of the IIV. However, all cohorts generated a vigorous, high-affinity, stalk-specific antibody response. Consistent increases in circulating memory B-cell frequencies after receipt of the IIV reflected the specificity of high-affinity antibody production. Our findings emphasize the value of LAIVs as a vehicle for prepandemic vaccination., (© The Author 2015. Published by Oxford University Press on behalf of the Infectious Diseases Society of America. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
22. Long-Lasting Impact of Neonatal Exposure to Total Body Gamma Radiation on Secondary Lymphoid Organ Structure and Function.
- Author
-
Rangel-Moreno J, de la Luz Garcia-Hernandez M, Ramos-Payan R, Biear J, Hernady E, Sangster MY, Randall TD, Johnston CJ, Finkelstein JN, and Williams JP
- Subjects
- Animals, Chemokines metabolism, Cytokines metabolism, Intercellular Signaling Peptides and Proteins metabolism, Lymphoid Tissue anatomy & histology, Lymphoid Tissue metabolism, Lymphoid Tissue physiology, Mice, Mice, Inbred C57BL, Animals, Newborn, Lymphoid Tissue radiation effects, Whole-Body Irradiation
- Abstract
The acute period after total body irradiation (TBI) is associated with an increased risk of infection, principally resulting from the loss of hematopoietic stem cells, as well as disruption of mucosal epithelial barriers. Although there is a return to baseline infection control coinciding with the apparent progressive recovery of hematopoietic cell populations, late susceptibility to infection in radiation-sensitive organs such as lung and kidney is known to occur. Indeed, pulmonary infections are particularly prevalent in hematopoietic cell transplant (HCT) survivors, in both adult and pediatric patient populations. Preclinical studies investigating late outcomes from localized thoracic irradiation have indicated that the mechanisms underlying pulmonary delayed effects are multifactorial, including exacerbated and persistent production of pro-inflammatory molecules and abnormal cross-talk among parenchymal and infiltrating immune and inflammatory cell populations. However, in the context of low-dose TBI, it is not clear whether the observed exacerbated response to infection remains contingent on these same mechanisms. It is possible instead, that after systemic radiation-induced injury, the susceptibility to infection may be independently related to defects in alternative organs that are revealed only through the challenge itself; indeed, we have hypothesized that this defect may be due to radiation-induced chronic effects in the structure and function of secondary lymphoid organs (SLO). In this study, we investigated the molecular and cellular alterations in SLO (i.e., spleen, mediastinal, inguinal and mesenteric lymph nodes) after TBI, and the time points when there appears to be immune competence. Furthermore, due to the high incidence of pulmonary infections in the late post-transplantation period of bone marrow transplant survivors, particularly in children, we focused on outcomes in mice irradiated as neonates, which served as a model for a pediatric population, and used the induction of adaptive immunity against influenza virus as a functional end point. We demonstrated that, in adult animals irradiated as neonates, high endothelial venule (HEV) expansion, generation of follicular helper T cells (TFH) and formation of splenic germinal centers (GC) were rapidly and, more importantly, persistently impaired in SLO, suggesting that the early-life exposure to sublethal radiation had long-lasting effects on the induction of humoral immunity. Although the neonatal TBI did not affect the overall outcome from influenza infection in the adults at the earlier time points assessed, we believe that they nonetheless contribute significantly to the increased mortality observed at subsequent late time points. Furthermore, we speculate that the detrimental and persistent impact on the induction of CD4 T- and B-cell responses in the mediastinal lymph nodes will decrease the animals' ability to respond to other aerial pathogens. Since many of these pathogens are normally cleared by antibodies, our findings provide an explanation for the susceptibility of survivors of childhood HCT to life-threatening respiratory tract infections. These findings have implications regarding the need for increased monitoring in pediatric hematopoietic cell transplant patients, since they indicate that there are ongoing and cumulative defects in SLO, which, importantly, develop during the immediate and early postirradiation period when patients may appear immunologically competent. The identification of changes in immune-related signals may offer bioindicators of progressive dysfunction, and of potential mechanisms that could be targeted so as to reduce the risk of infection from extracellular pathogens. Furthermore, these results support the potential susceptibility of the pediatric population to infection after sublethal irradiation in the context of a nuclear or radiological event.
- Published
- 2015
- Full Text
- View/download PDF
23. Robust mucosal-homing antibody-secreting B cell responses induced by intramuscular administration of adjuvanted bivalent human norovirus-like particle vaccine.
- Author
-
Sundararajan A, Sangster MY, Frey S, Atmar RL, Chen WH, Ferreira J, Bargatze R, Mendelman PM, Treanor JJ, and Topham DJ
- Subjects
- Adolescent, Adult, Animals, Double-Blind Method, Female, Humans, Immunization methods, Immunoglobulin A immunology, Immunoglobulin G immunology, Injections, Intramuscular, Male, Middle Aged, Placebos administration & dosage, Treatment Outcome, Vaccines, Virus-Like Particle administration & dosage, Vaccines, Virus-Like Particle genetics, Viral Vaccines administration & dosage, Viral Vaccines genetics, Young Adult, Adjuvants, Immunologic administration & dosage, Antibodies, Viral immunology, B-Lymphocytes immunology, Immunity, Mucosal, Norovirus immunology, Vaccines, Virus-Like Particle immunology, Viral Vaccines immunology
- Abstract
Background: Two major antigenically heterogenous norovirus genogroups (GI and GII) commonly infect humans and are the leading cause of foodborne, viral gastrointestinal infections in adults., Methods: We assessed B cell responses in participants in a double-blind, placebo-controlled, dose-escalation phase 1 study of the safety and immunogenicity of an intramuscular bivalent norovirus virus-like particle (VLP) vaccine. The vaccine contained a GI.1 VLP (Norwalk) and a consensus GII.4 VLP, representing the two major genotypes that cause human disease, and was administered on days 0 and 28 to healthy adults aged 18-49 years. Four separate cohorts received increasing doses of 5 μg, 15 μg, 50 μg, and 150 μg of each VLP adjuvanted in monophosphoryl lipid A and alum. PBMCs were analyzed for B cell activation and mucosal homing markers (flow cytometry) and VLP-specific and total IgG and IgA Ab-secreting cells (ASCs); and serum titers of VLP-specific IgG, IgA, and Pan-Ig were determined., Results: The vaccine elicited CD27+ CD38+ plasmablasts and high frequencies of ASCs specific for both VLP antigens in the peripheral blood at 7 days after the first dose. The plasmablasts exhibited a mucosal-homing phenotype and included a high proportion of IgA ASCs. Serum antibodies increased as early as 7 days after the first immunization., Conclusions: The data suggest that a single dose of the IM bivalent norovirus vaccine is effective in activating pre-existing B cell memory. The rapid B cell response and the mucosal homing phenotype of induced ASCs are consistent with anamnestic responses in subjects primed by prior oral norovirus infection. This study is registered at ClinicalTrials.gov Identifier NCT01609257., (Copyright © 2014 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
24. Live attenuated H7N7 influenza vaccine primes for a vigorous antibody response to inactivated H7N7 influenza vaccine.
- Author
-
Babu TM, Levine M, Fitzgerald T, Luke C, Sangster MY, Jin H, Topham D, Katz J, Treanor J, and Subbarao K
- Subjects
- Administration, Intranasal, Adult, B-Lymphocytes immunology, Enzyme-Linked Immunosorbent Assay, Enzyme-Linked Immunospot Assay, Flow Cytometry, Healthy Volunteers, Hemagglutination Inhibition Tests, Humans, Influenza A Virus, H7N3 Subtype immunology, Influenza Vaccines administration & dosage, Influenza, Human virology, Neutralization Tests, Real-Time Polymerase Chain Reaction, Vaccines, Attenuated administration & dosage, Vaccines, Attenuated immunology, Vaccines, Inactivated administration & dosage, Vaccines, Inactivated immunology, Virus Cultivation, Virus Shedding, Antibodies, Viral blood, Influenza A Virus, H7N7 Subtype immunology, Influenza Vaccines immunology, Influenza, Human prevention & control, Vaccination methods
- Abstract
Background: H7 influenza viruses have emerged as potential pandemic threat. We evaluated the safety and immunogenicity of two candidate H7 pandemic live attenuated influenza vaccines (pLAIV) and their ability to prime for responses to an unadjuvanted H7 pandemic inactivated influenza vaccine (pIIV)., Methods: Healthy seronegative adults received two doses of A/Netherlands/219/03 (H7N7) or one dose of A/chicken/British Columbia/CN-6/04 (H7N3) pLAIV all given as 10(7.5) 50% tissue culture infective doses (TCID50) intranasally. A subset of subjects received one 45 μg dose of H7N7 pIIV containing the A/Mallard/Netherlands/12/2000 HA intramuscularly 18-24 months after pLAIV. Viral shedding was assessed by culture and real-time polymerase chain reaction (rRT-PCR), B cell responses following pLAIV were evaluated by ELISPOT and flow cytometry. Serum antibody was assessed by hemagglutination-inhibition (HAI), microneutralization (MN) and ELISA assays after each vaccine., Results: Serum HAI or MN responses were not detected in any subject following one or two doses of either H7 pLAIV, although some subjects had detectable H7 specific B cells after vaccination. However, 10/13 subjects primed with two doses of H7N7 pLAIV responded to a subsequent dose of the homologous H7N7 pIIV with high titer HAI and MN antibody that cross-reacted with both North American and Eurasian lineage H7 viruses, including H7N9. In contrast, naïve subjects and recipients of a single dose of the mismatched H7N3 pLAIV did not develop HAI or MN antibody after pIIV., Conclusions: While pLAIVs did not elicit detectable serum MN or HAI antibody, strain-specific pLAIV priming established long term immune memory that was cross-reactive with other H7 influenza strains. Understanding the mechanisms underlying priming by pLAIV may aid in pandemic vaccine development., (Copyright © 2014. Published by Elsevier Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
25. Modeling the dynamics and migratory pathways of virus-specific antibody-secreting cell populations in primary influenza infection.
- Author
-
Miao H, Sangster MY, Livingstone AM, Hilchey SP, Zhang L, Topham DJ, Mosmann TR, Holden-Wiltse J, Perelson AS, Wu H, and Zand MS
- Subjects
- Animals, Antibody-Producing Cells cytology, Antibody-Producing Cells virology, Cell Movement, Immunoglobulin A immunology, Immunoglobulin G immunology, Immunoglobulin M immunology, Lung immunology, Lung virology, Lymph Nodes immunology, Lymph Nodes virology, Mice immunology, Mice, Inbred C57BL, Orthomyxoviridae Infections veterinary, Spleen immunology, Spleen virology, Antibodies, Viral immunology, Antibody-Producing Cells immunology, Computer Simulation, Mice virology, Models, Immunological, Orthomyxoviridae immunology, Orthomyxoviridae Infections immunology
- Abstract
The B cell response to influenza infection of the respiratory tract contributes to viral clearance and establishes profound resistance to reinfection by related viruses. Numerous studies have measured virus-specific antibody-secreting cell (ASC) frequencies in different anatomical compartments after influenza infection and provided a general picture of the kinetics of ASC formation and dispersion. However, the dynamics of ASC populations are difficult to determine experimentally and have received little attention. Here, we applied mathematical modeling to investigate the dynamics of ASC growth, death, and migration over the 2-week period following primary influenza infection in mice. Experimental data for model fitting came from high frequency measurements of virus-specific IgM, IgG, and IgA ASCs in the mediastinal lymph node (MLN), spleen, and lung. Model construction was based on a set of assumptions about ASC gain and loss from the sampled sites, and also on the directionality of ASC trafficking pathways. Most notably, modeling results suggest that differences in ASC fate and trafficking patterns reflect the site of formation and the expressed antibody class. Essentially all early IgA ASCs in the MLN migrated to spleen or lung, whereas cell death was likely the major reason for IgM and IgG ASC loss from the MLN. In contrast, the spleen contributed most of the IgM and IgG ASCs that migrated to the lung, but essentially none of the IgA ASCs. This finding points to a critical role for regional lymph nodes such as the MLN in the rapid generation of IgA ASCs that seed the lung. Results for the MLN also suggest that ASC death is a significant early feature of the B cell response. Overall, our analysis is consistent with accepted concepts in many regards, but it also indicates novel features of the B cell response to influenza that warrant further investigation.
- Published
- 2014
- Full Text
- View/download PDF
26. Passive broad-spectrum influenza immunoprophylaxis.
- Author
-
Berry CM, Penhale WJ, and Sangster MY
- Abstract
Influenza is a perennial problem affecting millions of people annually with the everpresent threat of devastating pandemics. Active prophylaxis by vaccination against influenza virus is currently the main countermeasure supplemented with antivirals. However, disadvantages of this strategy include the impact of antigenic drift, necessitating constant updating of vaccine strain composition, and emerging antiviral drug resistance. The development of other options for influenza prophylaxis, particularly with broad acting agents able to provide protection in the period between the onset of a pandemic and the development of a strain specific vaccine, is of great interest. Exploitation of broad-spectrum mediators could provide barricade protection in the early critical phase of influenza virus outbreaks. Passive immunity has the potential to provide immediate antiviral effects, inhibiting virus replication, reducing virus shedding, and thereby protecting vulnerable populations in the event of an impending influenza pandemic. Here, we review passive broad-spectrum influenza prophylaxis options with a focus on harnessing natural host defenses, including interferons and antibodies.
- Published
- 2014
- Full Text
- View/download PDF
27. CD4 T cell help is limiting and selective during the primary B cell response to influenza virus infection.
- Author
-
Alam S, Knowlden ZA, Sangster MY, and Sant AJ
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Viral blood, Enzyme-Linked Immunosorbent Assay, Immunodominant Epitopes chemistry, Immunodominant Epitopes immunology, Immunologic Memory, Mice, Molecular Sequence Data, Peptides chemistry, Peptides immunology, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, Influenza A virus isolation & purification, Orthomyxoviridae Infections immunology
- Abstract
Influenza virus vaccination strategies are focused upon the elicitation of protective antibody responses through administration of viral protein through either inactivated virions or live attenuated virus. Often overlooked in this strategy is the CD4 T cell response: how it develops into memory, and how it may support future primary B cell responses to heterologous infection. Through the utilization of a peptide-priming regimen, this study describes a strategy for developing CD4 T cell memory with the capacity to robustly expand in the lung-draining lymph node after live influenza virus infection. Not only were frequencies of antigen-specific CD4 T cells enhanced, but these cells also supported an accelerated primary B cell response to influenza virus-derived protein, evidenced by high anti-nucleoprotein (NP) serum antibody titers early, while there is still active viral replication ongoing in the lung. NP-specific antibody-secreting cells and heightened frequencies of germinal center B cells and follicular T helper cells were also readily detectable in the draining lymph node. Surprisingly, a boosted memory CD4 T cell response was not sufficient to provide intermolecular help for antibody responses. Our study demonstrates that CD4 T cell help is selective and limiting to the primary antibody response to influenza virus infection and that preemptive priming of CD4 T cell help can promote effective and rapid conversion of naive B cells to mature antibody-secreting cells.
- Published
- 2014
- Full Text
- View/download PDF
28. B cell response and hemagglutinin stalk-reactive antibody production in different age cohorts following 2009 H1N1 influenza virus vaccination.
- Author
-
Sangster MY, Baer J, Santiago FW, Fitzgerald T, Ilyushina NA, Sundararajan A, Henn AD, Krammer F, Yang H, Luke CJ, Zand MS, Wright PF, Treanor JJ, Topham DJ, and Subbarao K
- Subjects
- Adolescent, Adult, Age Factors, Aged, Aged, 80 and over, Animals, Cohort Studies, Female, Hemagglutination Inhibition Tests, Humans, Influenza Vaccines administration & dosage, Injections, Intramuscular, Male, Middle Aged, Neutralization Tests, Vaccines, Inactivated administration & dosage, Vaccines, Inactivated immunology, Young Adult, Antibodies, Neutralizing blood, Antibodies, Viral blood, B-Lymphocytes immunology, Hemagglutinin Glycoproteins, Influenza Virus immunology, Influenza A Virus, H1N1 Subtype immunology, Influenza Vaccines immunology
- Abstract
The 2009 pandemic H1N1 (pH1N1) influenza virus carried a swine-origin hemagglutinin (HA) that was closely related to the HAs of pre-1947 H1N1 viruses but highly divergent from the HAs of recently circulating H1N1 strains. Consequently, prior exposure to pH1N1-like viruses was mostly limited to individuals over the age of about 60 years. We related age and associated differences in immune history to the B cell response to an inactivated monovalent pH1N1 vaccine given intramuscularly to subjects in three age cohorts: 18 to 32 years, 60 to 69 years, and ≥70 years. The day 0 pH1N1-specific hemagglutination inhibition (HAI) and microneutralization (MN) titers were generally higher in the older cohorts, consistent with greater prevaccination exposure to pH1N1-like viruses. Most subjects in each cohort responded well to vaccination, with early formation of circulating virus-specific antibody (Ab)-secreting cells and ≥4-fold increases in HAI and MN titers. However, the response was strongest in the 18- to 32-year cohort. Circulating levels of HA stalk-reactive Abs were increased after vaccination, especially in the 18- to 32-year cohort, raising the possibility of elevated levels of cross-reactive neutralizing Abs. In the young cohort, an increase in MN activity against the seasonal influenza virus A/Brisbane/59/07 after vaccination was generally associated with an increase in the anti-Brisbane/59/07 HAI titer, suggesting an effect mediated primarily by HA head-reactive rather than stalk-reactive Abs. Our findings support recent proposals that immunization with a relatively novel HA favors the induction of Abs against conserved epitopes. They also emphasize the need to clarify how the level of circulating stalk-reactive Abs relates to resistance to influenza.
- Published
- 2013
- Full Text
- View/download PDF
29. Heterovariant cross-reactive B-cell responses induced by the 2009 pandemic influenza virus A subtype H1N1 vaccine.
- Author
-
He XS, Sasaki S, Baer J, Khurana S, Golding H, Treanor JJ, Topham DJ, Sangster MY, Jin H, Dekker CL, Subbarao K, and Greenberg HB
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Cross Reactions, Female, Hemagglutinin Glycoproteins, Influenza Virus immunology, Humans, Influenza, Human immunology, Influenza, Human virology, Male, Pandemics, Seasons, Young Adult, Antibodies, Viral blood, B-Lymphocytes immunology, Influenza A Virus, H1N1 Subtype immunology, Influenza Vaccines immunology
- Abstract
Background: The generation of heterovariant immunity is a highly desirable feature of influenza vaccines. The goal of this study was to compare the heterovariant B-cell response induced by the monovalent inactivated 2009 pandemic influenza A virus subtype H1N1 (A[H1N1]pdm09) vaccine with that induced by the 2009 seasonal trivalent influenza vaccine (sTIV) containing a seasonal influenza A virus subtype H1N1 (A[H1N1]) component in young and elderly adults., Methods: Plasmablast-derived polyclonal antibodies (PPAb) from young and elderly recipients of A(H1N1)pdm09 vaccine or sTIV were tested for binding activity to various influenza antigens., Results: In A(H1N1)pdm09 recipients, the PPAb titers against homotypic A(H1N1)pdm09 vaccine were similar to those against the heterovariant seasonal A(H1N1) vaccine and were similar between young and elderly subjects. The PPAb avidity was higher among elderly individuals, compared with young individuals. In contrast, the young sTIV recipients had 10-fold lower heterovariant PPAb titers against the A(H1N1)pdm09 vaccine than against the homotypic seasonal A(H1N1) vaccine. In binding assays with recombinant head and stalk domains of hemagglutinin, PPAb from the A(H1N1)pdm09 recipients but not PPAb from the sTIV recipients bound to the conserved stalk domain., Conclusion: The A(H1N1)pdm09 vaccine induced production of PPAb with heterovariant reactivity, including antibodies targeting the conserved hemagglutinin stalk domain.
- Published
- 2013
- Full Text
- View/download PDF
30. Host differences in influenza-specific CD4 T cell and B cell responses are modulated by viral strain and route of immunization.
- Author
-
Sundararajan A, Huan L, Richards KA, Marcelin G, Alam S, Joo H, Yang H, Webby RJ, Topham DJ, Sant AJ, and Sangster MY
- Subjects
- Animals, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, Influenza Vaccines immunology
- Abstract
The antibody response to influenza infection is largely dependent on CD4 T cell help for B cells. Cognate signals and secreted factors provided by CD4 T cells drive B cell activation and regulate antibody isotype switching for optimal antiviral activity. Recently, we analyzed HLA-DR1 transgenic (DR1) mice and C57BL/10 (B10) mice after infection with influenza virus A/New Caledonia/20/99 (NC) and defined epitopes recognized by virus-specific CD4 T cells. Using this information in the current study, we demonstrate that the pattern of secretion of IL-2, IFN-γ, and IL-4 by CD4 T cells activated by NC infection is largely independent of epitope specificity and the magnitude of the epitope-specific response. Interestingly, however, the characteristics of the virus-specific CD4 T cell and the B cell response to NC infection differed in DR1 and B10 mice. The response in B10 mice featured predominantly IFN-γ-secreting CD4 T cells and strong IgG2b/IgG2c production. In contrast, in DR1 mice most CD4 T cells secreted IL-2 and IgG production was IgG1-biased. Infection of DR1 mice with influenza PR8 generated a response that was comparable to that in B10 mice, with predominantly IFN-γ-secreting CD4 T cells and greater numbers of IgG2c than IgG1 antibody-secreting cells. The response to intramuscular vaccination with inactivated NC was similar in DR1 and B10 mice; the majority of CD4 T cells secreted IL-2 and most IgG antibody-secreting cells produced IgG2b or IgG2c. Our findings identify inherent host influences on characteristics of the virus-specific CD4 T cell and B cell responses that are restricted to the lung environment. Furthermore, we show that these host influences are substantially modulated by the type of infecting virus via the early induction of innate factors. Our findings emphasize the importance of immunization strategy for demonstrating inherent host differences in CD4 T cell and B cell responses.
- Published
- 2012
- Full Text
- View/download PDF
31. T cell immunoglobulin and mucin protein-3 (Tim-3)/Galectin-9 interaction regulates influenza A virus-specific humoral and CD8 T-cell responses.
- Author
-
Sharma S, Sundararajan A, Suryawanshi A, Kumar N, Veiga-Parga T, Kuchroo VK, Thomas PG, Sangster MY, and Rouse BT
- Subjects
- Acute-Phase Reaction immunology, Analysis of Variance, Animals, Enzyme-Linked Immunosorbent Assay, Female, Galectins genetics, Hepatitis A Virus Cellular Receptor 2, Mice, Mice, Inbred C57BL, Mice, Knockout, CD8-Positive T-Lymphocytes immunology, Galectins immunology, Immunity, Humoral immunology, Influenza A virus immunology, Orthomyxoviridae Infections immunology, Receptors, Virus immunology, Signal Transduction immunology
- Abstract
Reactions to pathogens are usually tuned to effect immunity and limit tissue damage. Several host counterinflammatory mechanisms inhibit tissue damage but these may also act to constrain the effectiveness of immunity to acute infections, as we demonstrate in mice acutely infected with influenza A virus (IAV). We show that compared with wild type (WT), galectin-9 knockout (G9KO) mice mounted a more robust acute phase virus-specific CD8 T-cell response as well as higher and more rapid virus-specific serum IgM, IgG, and IgA responses and also cleared virus more rapidly than did WT mice. Blocking galectin-9 signals to Tim-3-expressing cells using a Tim-3 fusion protein resulted in improved immune responses in WT mice. When IAV immune mice were challenged with a heterologous IAV, the secondary IAV-specific CD8 T-cell responses were four- to fivefold higher in G9KO compared with WT mice. Our results indicate that manipulating galectin signals may represent a convenient approach to improve immune responses to some vaccines.
- Published
- 2011
- Full Text
- View/download PDF
32. Time-dependent effects of pomegranate juice and pomegranate polyphenols on foodborne viral reduction.
- Author
-
Su X, Sangster MY, and D'Souza DH
- Subjects
- Animals, Calicivirus, Feline pathogenicity, Cats, Cell Line, Escherichia coli, Foodborne Diseases prevention & control, Foodborne Diseases virology, Humans, Hydrogen-Ion Concentration, Levivirus drug effects, Levivirus pathogenicity, Mice, Norovirus pathogenicity, Time Factors, Viral Load, Viral Plaque Assay, Antiviral Agents pharmacology, Beverages, Calicivirus, Feline drug effects, Lythraceae chemistry, Norovirus drug effects, Polyphenols pharmacology
- Abstract
Pomegranate juice (PJ) and pomegranate polyphenolic extracts (PP) have antiviral effects against HIV-1, influenza, herpes, and poxviruses, and we recently demonstrated their effect against human noroviral surrogates. In the present study, the time-dependent effects of two commercial brands of PJ and PP at two concentrations (2 and 4 mg/mL) on the infectivity of foodborne viral surrogates (feline calicivirus FCV-F9, murine norovirus MNV-1, and MS2 bacteriophage) at room temperature for up to 1 h were evaluated. Each virus at ∼5 log(10) plaque-forming units (PFU)/mL was mixed with equal volumes of PJ, or PP at 4 or 8 mg/mL, and incubated for 0, 10, 20, 30, 45, and 60 min at room temperature. Viral titers after each treatment were determined by standardized plaque assays and compared with untreated controls. Virus titer reduction by PJ and PP was found to be a rather rapid process, with ≥50% of titer reduction occurring within the first 20 min of treatment for all three tested viruses. Within the first 20 min, titer reductions of 3.12, 0.79, and 0.23 log(10) PFU/mL for FCV-F9, MNV-1, and MS2, respectively, were obtained using PJ. FCV-F9, MNV-1, and MS2 titers were reduced by 4.02, 0.68, and 0.18 log(10) PFU/mL with 2 mg/mL PP and 5.09, 1.14, and 0.19 log(10) PFU/mL with 4 mg/mL PP, respectively, after 20 min. The mechanism of viral reduction by PJ and PP needs to be elucidated and clinical trials should be undertaken before recommending for therapeutic or preventive purposes.
- Published
- 2011
- Full Text
- View/download PDF
33. In vitro effects of pomegranate juice and pomegranate polyphenols on foodborne viral surrogates.
- Author
-
Su X, Sangster MY, and D'Souza DH
- Subjects
- Animals, Calicivirus, Feline isolation & purification, Calicivirus, Feline pathogenicity, Cats, Cell Line, Foodborne Diseases prevention & control, Mice, Norovirus isolation & purification, Norovirus pathogenicity, Polyphenols, Viral Plaque Assay, Antiviral Agents pharmacology, Flavonoids pharmacology, Lythraceae chemistry, Phenols pharmacology
- Abstract
Pomegranate juice (PJ) has gained popularity because of its associated antioxidant, antimicrobial, anticancer, and anti-inflammatory properties. However, its effects against epidemiologically significant foodborne viruses have not been investigated. In the absence of culturable human noroviruses, feline calicivirus (FCV-F9), murine norovirus (MNV-1), and MS2 (ssRNA) bacteriophage were used as foodborne viral surrogates. The aim of this research was to study the effects of PJ and pomegranate polyphenols (PP) on foodborne viral infectivity. Viruses at high (∼ 7 log(10) PFU/mL) or low (∼ 5 log(10) PFU/mL) titers were mixed with equal volumes of PJ, 8, 16, and 32 mg/mL of PP, or water (control) and incubated for 1 h at room temperature. Viral infectivity after treatments was evaluated using standardized plaque assays. PJ decreased the titer of FCV-F9, MNV-1, and MS2 by 2.56, 1.32, and 0.32 log(10) PFU/mL, respectively, for low titers and 1.20, 0.06, and 0.63 log(10) PFU/mL, respectively, for high titers. Interestingly, FCV-F9 was undetectable after exposure to the three tested PP solutions using both low and high titers. MNV-1 at low initial titers was reduced by 1.30, 2.11, and 3.61 log(10) PFU/mL and at high initial titers by 1.56, 1.48, and 1.54 log(10) PFU/mL with 4, 8, and 16 mg/mL of PP treatment, respectively. MS2 at low initial titers was reduced by 0.41, 0.45, and 0.93 log(10) PFU/mL and at high initial titers by 0.32, 0.41, and 0.72 log(10) PFU/mL after 4, 8, and 16 mg/mL of PP treatment, respectively. PJ and PP resulted in titer reductions of foodborne virus surrogates after 1 h exposure, showing promise for use in hurdle technologies and/or for therapeutic or preventive use. To suggest the use of PJ and PP as natural remedies for foodborne viral illness prevention, their mechanism of action against viral infectivity needs to be further investigated.
- Published
- 2010
- Full Text
- View/download PDF
34. Influenza virus variation in susceptibility to inactivation by pomegranate polyphenols is determined by envelope glycoproteins.
- Author
-
Sundararajan A, Ganapathy R, Huan L, Dunlap JR, Webby RJ, Kotwal GJ, and Sangster MY
- Subjects
- Animals, Cell Line, Coronavirus drug effects, Enzyme-Linked Immunosorbent Assay, Hemagglutination, Viral drug effects, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Humans, Influenza A Virus, H1N1 Subtype drug effects, Influenza A Virus, H3N2 Subtype drug effects, Influenza A Virus, H5N1 Subtype drug effects, Influenza A virus physiology, Microbial Sensitivity Tests, Plant Extracts pharmacology, Polyphenols, Virus Inactivation drug effects, Antiviral Agents pharmacology, Flavonoids pharmacology, Influenza A virus drug effects, Lythraceae, Phenols pharmacology, Viral Envelope Proteins metabolism, Virus Attachment drug effects
- Abstract
Pomegranates have high levels of polyphenols (PPs) and may be a rich source of compounds with antiviral activity. We evaluated the direct anti-influenza activity of three commercially available pomegranate extracts: pomegranate juice (PJ), a concentrated liquid extract (POMxl), and a 93% PP powder extract (POMxp). The acidity of PJ and POMxl solutions contributed to rapid anti-influenza activity, but this was not a factor with POMxp. Studies using POMxp showed that 5min treatment at room temperature with 800μg/ml PPs resulted in at least a 3log reduction in the titers of influenza viruses PR8 (H1N1), X31 (H3N2), and a reassortant H5N1 virus derived from a human isolate. However, the antiviral activity was less against a coronavirus and reassortant H5N1 influenza viruses derived from avian isolates. The loss of influenza infectivity was frequently accompanied by loss of hemagglutinating activity. PP treatment decreased Ab binding to viral surface molecules, suggesting some coating of particles, but this did not always correlate with loss of infectivity. Electron microscopic analysis indicated that viral inactivation by PPs was primarily a consequence of virion structural damage. Our findings demonstrate that the direct anti-influenza activity of pomegranate PPs is substantially modulated by small changes in envelope glycoproteins., (Copyright © 2010 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
35. Quantitative analysis of influenza virus-specific B cell memory generated by different routes of inactivated virus vaccination.
- Author
-
Joo HM, He Y, Sundararajan A, Huan L, and Sangster MY
- Subjects
- Administration, Intranasal, Animals, Bone Marrow immunology, Injections, Intramuscular, Lung cytology, Lung immunology, Mice, Mice, Inbred C57BL, Vaccines, Inactivated administration & dosage, Vaccines, Inactivated immunology, Antibody-Producing Cells immunology, Immunologic Memory, Influenza Vaccines administration & dosage, Influenza Vaccines immunology, Vaccination methods
- Abstract
We consider both Ab-secreting cell (ASC) and memory B cell (B(Mem)) populations in a quantitative analysis of virus-specific B cell memory generated by intramuscular or intranasal vaccination of mice with inactivated influenza virus. After both forms of vaccination, the memory phase was characterized by localization of ASCs in the bone marrow and dispersion of B(Mem) to organized lymphoid tissues. The stronger IgG response to intramuscular vaccination correlated with larger numbers of IgG ASCs in the bone marrow and IgG B(Mem). IgA production was only prominent in the response to intranasal vaccination and was associated with IgA ASC localization in the lung and IgA B(Mem) formation. Notably, few IgG ASCs or B(Mem) localized in the lung after intramuscular vaccination, in contrast to the situation following influenza pneumonia. Our analysis links the nature of immunization to characteristics of the state of B cell memory that may relate to protective immunity., (Copyright 2009 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
36. Emu-BCL10 mice exhibit constitutive activation of both canonical and noncanonical NF-kappaB pathways generating marginal zone (MZ) B-cell expansion as a precursor to splenic MZ lymphoma.
- Author
-
Li Z, Wang H, Xue L, Shin DM, Roopenian D, Xu W, Qi CF, Sangster MY, Orihuela CJ, Tuomanen E, Rehg JE, Cui X, Zhang Q, Morse HC 3rd, and Morris SW
- Subjects
- Animals, B-Cell CLL-Lymphoma 10 Protein, B-Lymphocyte Subsets immunology, B-Lymphocyte Subsets metabolism, B-Lymphocyte Subsets pathology, Cell Proliferation, Cell Survival, Humans, Immunity, Humoral, Lymphoma, B-Cell, Marginal Zone genetics, Lymphoma, B-Cell, Marginal Zone metabolism, Lymphoma, B-Cell, Marginal Zone pathology, Mice, Mice, Transgenic, Recombinant Proteins genetics, Recombinant Proteins metabolism, Signal Transduction, Splenic Neoplasms genetics, Splenic Neoplasms metabolism, Splenic Neoplasms pathology, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Lymphoma, B-Cell, Marginal Zone etiology, NF-kappa B metabolism, Splenic Neoplasms etiology
- Abstract
BCL10, required for nuclear factor kappaB (NF-kappaB) activation during antigen-driven lymphocyte responses, is aberrantly expressed in mucosa-associated lymphoid tissue-type marginal zone (MZ) lymphomas because of chromosomal translocations. Emu-driven human BCL10 transgenic (Tg) mice, which we created and characterize here, had expanded populations of MZ B cells and reduced follicular and B1a cells. Splenic B cells from Tg mice exhibited constitutive activation of both canonical and noncanonical NF-kappaB signaling pathways is associated with increased expression of NF-kappaB target genes. These genes included Tnfsf13b, which encodes the B-cell activating factor (BAFF). In addition, levels of BAFF were significantly increased in sera from Tg mice. MZ B cells of Tg mice exhibited reduced turnover in vivo and enhanced survival in vitro, indicative of lymphoaccumulation rather than lymphoproliferation as the cause of MZ expansion. In vivo antibody responses to both T-independent, and especially T-dependent, antigens were significantly reduced in Tg mice. Mortality was accelerated in Tg animals, and some mice older than 8 months had histologic and molecular findings indicative of clonal splenic MZ lymphoma. These results suggest that, in addition to constitutive activation of BCL10 in MZ B cells, other genetic factors or environmental influences are required for short latency oncogenic transformation.
- Published
- 2009
- Full Text
- View/download PDF
37. The generation of influenza-specific humoral responses is impaired in ST6Gal I-deficient mice.
- Author
-
Zeng J, Joo HM, Rajini B, Wrammert JP, Sangster MY, and Onami TM
- Subjects
- Animals, Antibody Formation immunology, B-Lymphocytes cytology, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Female, Germinal Center immunology, Immunoglobulin M immunology, Immunologic Memory immunology, Lymphocyte Activation immunology, Mice, Mice, Knockout, Sialyltransferases genetics, Virus Replication, beta-D-Galactoside alpha 2-6-Sialyltransferase, Immunity, Innate immunology, Influenza A virus immunology, Sialyltransferases deficiency, Sialyltransferases metabolism
- Abstract
Posttranslational modification of proteins, such as glycosylation, can impact cell signaling and function. ST6Gal I, a glycosyltransferase expressed by B cells, catalyzes the addition of alpha-2,6 sialic acid to galactose, a modification found on N-linked glycoproteins such as CD22, a negative regulator of B cell activation. We show that SNA lectin, which binds alpha-2,6 sialic acid linked to galactose, shows high binding on plasma blasts and germinal center B cells following viral infection, suggesting ST6Gal I expression remains high on activated B cells in vivo. To understand the relevance of this modification on the antiviral B cell immune response, we infected ST6Gal I(-/-) mice with influenza A/HKx31. We demonstrate that the loss of ST6Gal I expression results in similar influenza infectivity in the lung, but significantly reduced early influenza-specific IgM and IgG levels in the serum, as well as significantly reduced numbers of early viral-specific Ab-secreting cells. At later memory time points, ST6Gal I(-/-) mice show comparable numbers of IgG influenza-specific memory B cells and long-lived plasma cells, with similarly high antiviral IgG titers, with the exception of IgG2c. Finally, we adoptively transfer purified B cells from wild-type or ST6Gal I(-/-) mice into B cell-deficient (microMT(-/-)) mice. Recipient mice that received ST6Gal I(-/-) B cells demonstrated reduced influenza-specific IgM levels, but similar levels of influenza-specific IgG, compared with mice that received wild-type B cells. These data suggest that a B cell intrinsic defect partially contributes to the impaired antiviral humoral response.
- Published
- 2009
- Full Text
- View/download PDF
38. Broad dispersion and lung localization of virus-specific memory B cells induced by influenza pneumonia.
- Author
-
Joo HM, He Y, and Sangster MY
- Subjects
- Animals, Female, Immunoglobulin A, Immunoglobulin G, Influenza A Virus, H3N2 Subtype immunology, Lung pathology, Lymphocyte Count, Lymphoid Tissue pathology, Mice, Tissue Distribution, B-Lymphocytes immunology, Immunologic Memory, Orthomyxoviridae Infections immunology, Pneumonia, Viral immunology
- Abstract
Although memory B cells (B(Mem)) contribute significantly to resistance to infection, B(Mem) population characteristics that may relate to protective efficacy have received little attention. Here, we report a comprehensive quantitative analysis of virus-specific IgG and IgA B(Mem) dispersion after transient influenza pneumonia in mice. From early in the response, B(Mem) circulated continuously and dispersed widely to secondary lymphoid tissues. However, a complicated picture emerged with B(Mem) frequency differences between secondary lymphoid tissues indicating an influence of local tissue factors on trafficking. B(Mem) numbers increased and stabilized at tissue-specific frequencies without contraction of the B(Mem) pool during the period of analysis. The lung was notable as a nonsecondary lymphoid tissue where a rapid influx of IgG and IgA B(Mem) established relatively high frequencies that were maintained long term. Our findings provide insights into the pattern of B(Mem) dispersion, and emphasize the lung as a complex repository of immune memory after local infection.
- Published
- 2008
- Full Text
- View/download PDF
39. Quantitative analysis of herpes simplex virus type 1-specific memory B cells generated by different routes of infection.
- Author
-
Vanitha DJ, Joo HM, Rouse BT, and Sangster MY
- Subjects
- Animals, Antibody Specificity, Herpes Simplex blood, Immunization, Secondary, Immunoglobulin G blood, Immunoglobulin G immunology, Immunologic Memory, Lymph Nodes immunology, Lymphocyte Count, Mediastinum, Mice, Mice, Inbred C57BL, Organ Specificity, Peyer's Patches immunology, Spleen immunology, B-Lymphocytes immunology, Herpes Simplex immunology, Herpesvirus 1, Human immunology, Immunization
- Abstract
We compared the herpes simplex virus type 1 (HSV)-specific memory B cell (MBC) populations generated by footpad and intranasal infection in mice. Both routes of infection generated transient antibody-secreting cell responses in the draining lymph nodes and spleen, and sustained circulating IgG. HSV-specific IgG MBCs, analyzed by limiting dilution assay approximately 8 weeks after infection, were distributed in a range of lymph nodes and in the spleen and Peyer's patches. Overall, the route of infection had little effect on the MBC frequency in each anatomical location. Interestingly, after both routes of infection there was a trend towards preferential MBC accumulation in the mediastinal lymph node. Intravaginal challenge of mice primed by footpad or intranasal infection generated similar secondary IgG responses. Our findings indicate that the widespread dispersion of MBCs to lymphoid tissues throughout the body is largely independent of the route of infection, but may be influenced by tissue-specific factors.
- Published
- 2007
- Full Text
- View/download PDF
40. A strategy for selective, CD4+ T cell-independent activation of virus-specific memory B cells for limiting dilution analysis.
- Author
-
Li X, Vanitha DJ, Joo HM, He Y, Rouse BT, and Sangster MY
- Subjects
- Animals, B-Lymphocytes cytology, Cell Count, Chlorocebus aethiops, Coculture Techniques, Female, Herpes Simplex immunology, Herpesvirus 1, Human immunology, Humans, Influenza A Virus, H3N2 Subtype chemistry, Influenza A Virus, H3N2 Subtype immunology, Lymphoid Tissue cytology, Lymphoid Tissue immunology, Mice, Mice, Inbred C57BL, Orthomyxoviridae Infections immunology, Spleen cytology, Spleen immunology, Spleen virology, Vero Cells, Viruses chemistry, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, Immunologic Memory immunology, Lymphocyte Activation immunology, Viruses immunology
- Abstract
Complete characterization of the B cell response to infection or vaccination is dependent on accurate quantitation of the memory B cell (MBC) pool. An established method for measuring MBC frequencies is limiting dilution analysis based on in vitro stimulation of MBCs to divide and differentiate into antibody-secreting cells (ASCs). The presence of specific antibody then serves to identify cultures positive for precursor MBCs. The sensitivity of this approach is critically dependent on optimal in vitro MBC activation. To develop a limiting dilution assay (LDA) for measuring influenza-specific MBC frequencies, we evaluated strategies for the in vitro stimulation of influenza-specific MBCs. An ELISPOT assay to enumerate influenza-specific IgG ASCs was used as the readout for MBC activation. Culture of influenza-specific MBCs with influenza-infected splenocytes was effective for MBC activation, but T cell-associated factors were required for optimal LDA sensitivity and clonal expansion of activated MBCs. However, optimal influenza-specific MBC activation was T cell-independent when MBCs were simply cultured with beta-propiolactone (BPL)-inactivated influenza virus particles (BPL-flu). BPL-flu did not stimulate naïve B cells to produce influenza-specific IgG, demonstrating that only MBCs were activated. In addition, BPL-flu acted selectively and only activated influenza-specific MBCs, not MBCs of other specificities. Analysis of influenza-specific MBC frequencies in different anatomical locations in influenza-immune mice established that in vitro stimulation with BPL-flu provided the basis for a sensitive and reproducible LDA. Extending our studies to the herpes simplex virus (HSV) system, we demonstrated that HSV-specific MBCs cultured with BPL-inactivated HSV were selectively activated to IgG secretion in the absence of T cells. Our studies identify BPL-inactivated viral particles as a valuable tool for selective, T cell-independent activation of virus-specific MBCs in vitro. This strategy eliminates the influence of poorly defined T cell-associated factors on MBC frequency determinations.
- Published
- 2006
- Full Text
- View/download PDF
41. Pathogenesis of Hong Kong H5N1 influenza virus NS gene reassortants in mice: the role of cytokines and B- and T-cell responses.
- Author
-
Lipatov AS, Andreansky S, Webby RJ, Hulse DJ, Rehg JE, Krauss S, Perez DR, Doherty PC, Webster RG, and Sangster MY
- Subjects
- Animals, Disease Models, Animal, Female, Humans, Influenza A virus genetics, Influenza, Human immunology, Influenza, Human physiopathology, Influenza, Human virology, Lung immunology, Lung physiopathology, Lung virology, Mice, Mice, Inbred BALB C, Reassortant Viruses genetics, Viral Nonstructural Proteins genetics, Virulence, B-Lymphocytes immunology, Cytokines biosynthesis, Influenza A Virus, H5N1 Subtype, Influenza A virus pathogenicity, Reassortant Viruses pathogenicity, T-Lymphocytes immunology, Viral Nonstructural Proteins immunology
- Abstract
The severity of disease caused in humans by H5N1 influenza viruses remains unexplained. The NS gene of Hong Kong H5N1/97 viruses was shown to contribute to high pathogenicity of reassortants in a pig model. However, the molecular pathogenesis and host immune response underlying this phenomenon remain unclear. Here, in a mouse model, H1N1 A/Puerto Rico/8/34 (PR/8) reassortants that contained the H5N1/97 NS gene, the H5N1/01 NS gene, or an altered H5N1/97 NS gene encoding a Glu92-->Asp substitution in NS1 was studied. The pathogenicity of reassortant viruses, the induction of cytokines and chemokine CXCL1 (KC) in the lungs and specific B- and T-cell responses was characterized. In mice infected with reassortant virus containing the H5N1/97 NS gene, the mouse lethal dose (50%) and lung virus titres were similar to those of PR/8, which is highly pathogenic to mice. This reassortant virus required two more days than PR/8 to be cleared from the lungs of infected mice. Reassortants containing the altered H5N1/97 NS gene or the H5N1/01 NS gene demonstrated attenuated pathogenicity and lower lung titres in mice. Specific B- and T-cell responses were consistent with viral pathogenicity and did not explain the delayed clearance of the H5N1/97 NS reassortant. The reassortant induced elevated pulmonary concentrations of the inflammatory cytokines IL1alpha, IL1beta, IL6, IFN-gamma and chemokine KC, and decreased concentrations of the anti-inflammatory cytokine IL10. This cytokine imbalance is reminiscent of the clinical findings in two humans who died of H5N1/97 infection and may explain the unusual severity of the disease.
- Published
- 2005
- Full Text
- View/download PDF
42. An early CD4+ T cell-dependent immunoglobulin A response to influenza infection in the absence of key cognate T-B interactions.
- Author
-
Sangster MY, Riberdy JM, Gonzalez M, Topham DJ, Baumgarth N, and Doherty PC
- Subjects
- Animals, CD40 Antigens physiology, Histocompatibility Antigens Class II physiology, Immunoglobulin G biosynthesis, Immunologic Memory, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Antibodies, Viral biosynthesis, B-Lymphocytes physiology, CD4-Positive T-Lymphocytes physiology, Cell Communication, Immunoglobulin A biosynthesis, Orthomyxoviridae Infections immunology, T-Lymphocytes physiology
- Abstract
Contact-mediated interactions between CD4+ T cells and B cells are considered crucial for T cell-dependent B cell responses. To investigate the ability of activated CD4+ T cells to drive in vivo B cell responses in the absence of key cognate T-B interactions, we constructed radiation bone marrow chimeras in which CD4+ T cells would be activated by wild-type (WT) dendritic cells, but would interact with B cells that lacked expression of either major histocompatibility complex class II (MHC II) or CD40. B cell responses were assessed after influenza virus infection of the respiratory tract, which elicits a vigorous, CD4+ T cell-dependent antibody response in WT mice. The influenza-specific antibody response was strongly reduced in MHC II knockout and CD40 knockout mice. MHC II-deficient and CD40-deficient B cells in the chimera environment also produced little virus-specific immunoglobulin (Ig)M and IgG, but generated a strong virus-specific IgA response with virus-neutralizing activity. The IgA response was entirely influenza specific, in contrast to the IgG2a response, which had a substantial nonvirus-specific component. Our study demonstrates a CD4+ T cell-dependent, antiviral IgA response that is generated in the absence of B cell signaling via MHC II or CD40, and is restricted exclusively to virus-specific B cells.
- Published
- 2003
- Full Text
- View/download PDF
43. Measuring the diaspora for virus-specific CD8+ T cells.
- Author
-
Marshall DR, Turner SJ, Belz GT, Wingo S, Andreansky S, Sangster MY, Riberdy JM, Liu T, Tan M, and Doherty PC
- Subjects
- Animals, Antigens, CD, Antigens, Differentiation, T-Lymphocyte, Female, H-2 Antigens immunology, Histocompatibility Antigen H-2D, Humans, Immunologic Memory immunology, Kinetics, Lectins, C-Type, Lymphoid Tissue immunology, Mice, Mice, Inbred C57BL, Peptides immunology, Respiratory Mucosa immunology, Tissue Distribution, Antigens, Viral immunology, CD8-Positive T-Lymphocytes immunology, DNA-Directed RNA Polymerases immunology, Influenza A virus immunology, Peptide Fragments immunology, RNA-Dependent RNA Polymerase, Viral Core Proteins immunology, Viral Proteins immunology
- Abstract
The CD8(+) T cell diaspora has been analyzed after secondary challenge with an influenza A virus that replicates only in the respiratory tract. Numbers of D(b)NP(366)- and D(b)PA(224)-specific CD8(+) T cells were measured by tetramer staining at the end of the recall response, then followed sequentially in the lung, lymph nodes, spleen, blood, and other organs. The extent of clonal expansion did not reflect the sizes of the preexisting memory T cell pools. Although the high-frequency CD8(+) tetramer(+) populations in the pneumonic lung and mediastinal lymph nodes fell rapidly from peak values, the "whole mouse" virus-specific CD8(+) T cell counts decreased only 2-fold over the 4 weeks after infection, then subsided at a fairly steady rate to reach a plateau at about 2 months. The largest numbers were found throughout in the spleen, then the bone marrow. The CD8(+)D(b)NP(366)+ and CD8(+)D(b)PA(224)+ sets remained significantly enlarged for at least 4 months, declining at equivalent rates while retaining the nucleoprotein > acid polymerase immunodominance hierarchy characteristic of the earlier antigen-driven phase. Lowest levels of the CD69 "activation marker" were detected consistently on virus-specific CD8(+) T cells in the blood, then the spleen. Those in the bone marrow and liver were intermediate, and CD69(hi) T cells were very prominent in the regional lymph nodes and the nasal-associated lymphoid tissue. Any population of "resting" CD8(+) memory T cells is thus phenotypically heterogeneous, widely dispersed, and subject to broad homeostatic and local environmental effects irrespective of epitope specificity or magnitude.
- Published
- 2001
- Full Text
- View/download PDF
44. Dissecting the host response to a gamma-herpesvirus.
- Author
-
Doherty PC, Christensen JP, Belz GT, Stevenson PG, and Sangster MY
- Subjects
- Animals, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Chemokines immunology, Cytokines immunology, Herpesviridae Infections pathology, Herpesviridae Infections prevention & control, Herpesviridae Infections virology, Humans, Immunization, Tumor Virus Infections pathology, Tumor Virus Infections prevention & control, Tumor Virus Infections virology, Gammaherpesvirinae immunology, Herpesviridae Infections immunology, Tumor Virus Infections immunology
- Abstract
The murine gamma-herpesvirus 68 (MHV-68) provides a unique experimental model for dissecting immunity to large DNA viruses that persist in B lymphocytes. The analysis is greatly facilitated by the availability of genetically disrupted (-/-) mice that lack key host-response elements, and by the fact that MHV-68 is a lytic virus that can readily be manipulated for mutational analysis. The mutant virus strategy is being used, for example, to characterize the part played in vivo by an MHV-68-encoded chemokine-binding protein that may ultimately find an application in human therapeutics. Experiments with various -/- mice and monoclonal antibody depletion protocols have shown very clearly that type I interferons (IFNs) are essential for the early control of MHV-68 replication, while CD4+ T cells producing IFN-gamma function to limit the consequences of viral persistence. Virus-specific CD8+ effectors acting in the absence of the CD4+ subset seem initially to control the lytic phase in the lung following respiratory challenge, but are then unable to prevent the reactivation of replicative infection in epithelia and the eventual death of CD4+ T-cell-deficient mice. This could reflect the fact that the interaction between the CD8+ T cells and the virus-infected targets is partially compromised by the MHV-68 K3 protein, which inhibits antigen presentation by MHC class I glycoproteins. Immunization strategies focusing on the CD8+ T-cell response to epitopes expressed during the lytic phase of MHV-68 infection can limit virus replication, but are unable to prevent the establishment of latency. Other experiments with mutant viruses also suggest that there is a disconnection between lytic MHV-68 infection and latency. The massive nonspecific immunoglobulin response and the dramatic expansion of Vbeta4+ CD8+ T cells, which is apparently MHC independent, could represent some sort of 'smoke screen' used by MHV-68 to subvert immunity. Although MHV-68 is neither Epstein-Barr virus nor human herpesvirus-8, the results generated from this system suggest possibilities that may usefully be addressed with these human pathogens. Perhaps the main lesson learned to date is that all the components of immunity are likely to be important for the control of these complex viruses.
- Published
- 2001
- Full Text
- View/download PDF
45. Phospholipase Cgamma2 is essential in the functions of B cell and several Fc receptors.
- Author
-
Wang D, Feng J, Wen R, Marine JC, Sangster MY, Parganas E, Hoffmeyer A, Jackson CW, Cleveland JL, Murray PJ, and Ihle JN
- Subjects
- Adaptor Proteins, Signal Transducing, Agammaglobulinaemia Tyrosine Kinase, Animals, B-Lymphocytes metabolism, Blood Platelets physiology, Carrier Proteins metabolism, Hematopoiesis physiology, Isoenzymes genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Phospholipase C gamma, Phosphoproteins metabolism, Protein-Tyrosine Kinases metabolism, Receptors, Antigen, B-Cell metabolism, Receptors, IgG metabolism, Type C Phospholipases genetics, B-Lymphocytes physiology, Isoenzymes physiology, Receptors, IgG physiology, Type C Phospholipases physiology
- Abstract
Many receptors activate phospholipase Cgamma1 or -gamma2. To assess the role of PLCgamma2, we derived enzyme-deficient mice. The mice are viable but have decreased mature B cells, a block in pro-B cell differentiation, and B1 B cell deficiency. IgM receptor-induced Ca2+ flux and proliferation to B cell mitogens are absent. IgM, IgG2a, and IgG3 levels are reduced, and T cell-independent antibody production is absent. The similarity to Btk- or Blnk-deficient mice demonstrates that PLCgamma2 is downstream in Btk/Blnk signaling. FcRgamma signaling is also defective, resulting in a loss of collagen-induced platelet aggregation, mast cell FcepsilonR function, and NK cell FcgammaRIII and 2B4 function. The results define a signal transduction pathway broadly utilized by immunoglobulin superfamily receptors.
- Published
- 2000
- Full Text
- View/download PDF
46. Analysis of the virus-specific and nonspecific B cell response to a persistent B-lymphotropic gammaherpesvirus.
- Author
-
Sangster MY, Topham DJ, D'Costa S, Cardin RD, Marion TN, Myers LK, and Doherty PC
- Subjects
- Animals, Antibodies, Viral biosynthesis, Antibodies, Viral blood, Antibody-Producing Cells metabolism, Autoantibodies biosynthesis, B-Lymphocytes metabolism, Bone Marrow Cells immunology, Bone Marrow Cells metabolism, Clone Cells, Cytokines deficiency, Cytokines genetics, Herpesviridae Infections blood, Kinetics, Lymph Nodes immunology, Lymph Nodes metabolism, Lymphocyte Activation, Lymphocyte Cooperation, Mice, Mice, Inbred C57BL, Mice, Knockout, Spleen immunology, Spleen metabolism, T-Lymphocytes immunology, Antibody Specificity, B-Lymphocytes immunology, B-Lymphocytes virology, Gammaherpesvirinae immunology, Herpesviridae Infections immunology
- Abstract
Respiratory challenge of mice with murine gammaherpesvirus 68 (gammaHV68) results in acute replication in respiratory epithelial cells and persistent, latent infection of B cells and macrophages. gammaHV68 elicits virus-specific Ab, and also nonspecifically activates B cells to Ab production through a CD4+ T cell-dependent process. The current analysis characterizes virus-specific and nonspecific Ab production at the single cell level and investigates the requirements and nature of the nonspecific response. Virus-specific Ab-forming cell (AFC) numbers were dwarfed by the increase in total AFC in all sites examined, indicating substantial nonspecific Ab production. Clear increases and decreases in specific and total AFC numbers occurred in the lymph nodes draining the respiratory tract and the spleen, but AFC numbers in the bone marrow (BM) increased to a plateau and remained constant. The longevity of the BM response was reflected in a sustained increase in virus-specific and total serum Ab levels. Generally, the IgG2a and IgG2b isotypes predominated. Analysis of cytokine-deficient mice, CD40 ligand-deficient mice, and radiation BM chimeras lacking MHC class II expression specifically on B cells indicated that nonspecific Ab production is independent of IL-6 or IFN-gamma, and dependent on cognate CD4+ T cell help. Several observations were consistent with polyclonal B cell activation by gammaHV68, including the induction of durable serum levels of IgG reactive with mammalian dsDNA and murine type II collagen. Our findings indicate new directions for studies of this valuable model of gamma-herpesvirus pathogenesis.
- Published
- 2000
- Full Text
- View/download PDF
47. Development and characterization of new flavivirus-resistant mouse strains bearing Flv(r)-like and Flv(mr) alleles from wild or wild-derived mice.
- Author
-
Urosevic N, Silvia OJ, Sangster MY, Mansfield JP, Hodgetts SI, and Shellam GR
- Subjects
- Animals, Genetic Predisposition to Disease, Mice, Mice, Inbred BALB C, Mice, Inbred C3H, Polymorphism, Genetic, Species Specificity, Virus Replication, Alleles, Chromosome Mapping, Flavivirus Infections genetics
- Abstract
A single genetic locus, flavivirus resistance (Flv), controls virus titres and severity of flavivirus infection in mouse brain. It has been mapped to mouse chromosome 5 and shown to include different allelic forms. While the majority of laboratory mouse strains are susceptible to flaviviruses and carry the Flv(s) allele, wild mice and laboratory mouse strains recently derived from them are resistant and carry flavivirus-resistance alleles including Flv(r)-like and Flv(mr) alleles. Although there is a mouse model of flavivirus resistance conferred by the Flv(r) allele, other resistance alleles have not been adequately studied due to a lack of appropriate animal models. In this paper we describe the development of new flavivirus-resistant mouse strains, C3H.M.domesticus-Flv(r) and C3H.MOLD-Flv(mr), which carry the novel resistance alleles Flv(r)-like and Flv(mr) on the genetic background of flavivirus susceptible C3H/HeJ mice. The new strains were created by 10 to 11 generations of backcrossing followed by brother-sister matings resulting in a generation of homozygous founder stocks. Genome analysis of the newly developed mouse strains has revealed chromosomal regions of approximately 9 and 11 cM, respectively, encompassing Flv on chromosome 5, which are derived from resistant donor mice. These segments are much smaller than the segment of approximately 31 cM described in the congenic resistant mouse strain C3H.PRI-Flv(r) (also known as C3H/RV). The new congenic mouse strains, which were created to carry the Flv(r)-like and Flv(mr) alleles on the standardized genetic background of susceptible mice, represent new animal models of flavivirus resistance conferred by these novel resistance alleles.
- Published
- 1999
- Full Text
- View/download PDF
48. Stat5 is required for IL-2-induced cell cycle progression of peripheral T cells.
- Author
-
Moriggl R, Topham DJ, Teglund S, Sexl V, McKay C, Wang D, Hoffmeyer A, van Deursen J, Sangster MY, Bunting KD, Grosveld GC, and Ihle JN
- Subjects
- Animals, Cell Division, Cells, Cultured, Flow Cytometry, Mice, Mice, Mutant Strains, STAT5 Transcription Factor, Thymus Gland cytology, Thymus Gland metabolism, Cell Cycle physiology, DNA-Binding Proteins physiology, Interleukin-2 physiology, Milk Proteins, T-Lymphocytes cytology, Trans-Activators physiology
- Abstract
Many cytokines activate two highly homologous Stat proteins, 5a and 5b. Mice deficient in both genes lack all growth hormone and prolactin functions but retain functions associated with cytokines such as erythropoietin. Here, we demonstrate that, while lymphoid development is normal, Stat5a/b mutant peripheral T cells are profoundly deficient in proliferation and fail to undergo cell cycle progression or to express genes controlling cell cycle progression. In addition, the mice lack NK cells, develop splenomegaly, and have T cells with an activated phenotype, phenotypes seen in IL-2 receptor beta chain-deficient mice. These phenotypes are not seen in mice lacking Stat5a or Stat5b alone. The results demonstrate that the Stat5 proteins, redundantly, are essential mediators of IL-2 signaling in T cells.
- Published
- 1999
- Full Text
- View/download PDF
49. Thymic lymphoproliferative disease after successful correction of CD40 ligand deficiency by gene transfer in mice.
- Author
-
Brown MP, Topham DJ, Sangster MY, Zhao J, Flynn KJ, Surman SL, Woodland DL, Doherty PC, Farr AG, Pattengale PK, and Brenner MK
- Subjects
- Animals, Bone Marrow Cells cytology, Bone Marrow Cells immunology, CD40 Ligand, Gene Transfer Techniques, Immunity, Cellular genetics, Lymphoma immunology, Lymphoproliferative Disorders immunology, Membrane Glycoproteins deficiency, Membrane Glycoproteins genetics, Mice, Mice, Knockout, Polymerase Chain Reaction, Retroviridae genetics, Retroviridae physiology, Thymus Gland immunology, Thymus Neoplasms immunology, Transduction, Genetic, Virus Replication, X Chromosome, Genetic Therapy, Lymphoproliferative Disorders genetics, Lymphoproliferative Disorders therapy, Membrane Glycoproteins physiology, T-Lymphocytes immunology
- Abstract
Inherited deficiency of the CD40 ligand (X-linked hyper-IgM syndrome) is characterized by failure of immunoglobulin isotype switching and severe defects of cell-mediated immunity. To test the potential for gene transfer therapy to correct this disorder, we transduced murine bone marrow or thymic cells with a retroviral vector containing the cDNA for the murine CD40 ligand (CD40L) and injected them into CD40L-/- mice. Even low-level, constitutive expression of the transgene stimulated humoral and cellular immune functions in these mice. With extended follow-up, however, 12 of 19 treated mice developed T-lymphoproliferative disorders, ranging from polyclonal increases of lymphoblasts to overt monoclonal T-lymphoblastic lymphomas that involved multiple organs. Our findings show that constitutive (rather than tightly regulated), low-level expression of CD40L can produce abnormal proliferative responses in developing T lymphocytes, apparently through aberrant interaction between CD40L+ and TCRalphabeta+CD40+ thymocytes. Current methods of gene therapy may prove inappropriate for disorders involving highly regulated genes in essential positions in proliferative cascades.
- Published
- 1998
- Full Text
- View/download PDF
50. Genetic control of host resistance to flavivirus infection in animals.
- Author
-
Shellam GR, Sangster MY, and Urosevic N
- Subjects
- Animals, Flavivirus Infections genetics, Flavivirus Infections immunology, Humans, Immunity, Innate genetics, Virus Replication, Animals, Domestic, Animals, Wild, Arthropod Vectors virology, Flavivirus physiology, Flavivirus Infections veterinary
- Abstract
Flaviviruses are small, enveloped RNA viruses which are generally transmitted by arthropods to animals and man. Although flaviviruses cause important diseases in domestic animals and man, flaviviral infection of animals which constitute the normal vertebrate reservoir may be mild or sub-clinical, which suggests that some adaptation between virus and host may have occurred. While this possibility is difficult to study in wild animals, extensive studies using laboratory mice have demonstrated the existence of innate, flavivirus-specific resistance. Resistance is heritable and is attributable to the gene Flvr, which is located on chromosome 5 in this species. The mechanism of resistance is at present unknown, but acts early and limits the replication of flaviviruses in cells. While some evidence supports a role for Flvr in enhancing the production of defective interfering virus, thereby restricting the production of infectious virus, other reports suggest that Flvr interferes with either virus RNA replication or RNA packaging. Recent research suggests that cytoplasmic proteins bind to the viral replication complex and that allelic forms of these proteins in resistant mice may restrict the production of infectious progeny. Apparent resistance to flaviviruses has been described in other vertebrates, although it remains to be seen if this is attributable to a homologue of Flvr. Nonetheless, knowledge gained of the characteristics and function of Flvr in mice should be applicable to other host species, and improvement of resistance to flaviviral infection in domestic animals by selective breeding or gene technology may ultimately be possible.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.