Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Publication year range
1.
Immun Ageing ; 20(1): 4, 2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36650551

ABSTRACT

BACKGROUND: Immune responses to COVID-19 mRNA vaccines have not been well characterized in frail older adults. We postulated that frailty is associated with impaired antibody and cellular mRNA vaccine responses. METHODS: We followed older adults in a retirement facility with longitudinal clinical and serological samples from the first Moderna mRNA-1273 vaccine dose starting in February 2021 through their 3rd (booster) vaccine dose. Outcomes were antibody titers, antibody avidity, and AIM+ T cell function and phenotype. Statistical analysis used linear regression with clustered error for antibody titers over multiple timepoints with clinical predictors including, age, sex, prior infection status, and clinical frailty scale (CFS) score. T cell function analysis used linear regression models with clinical predictors and cellular memory phenotype variables. RESULTS: Participants (n = 15) had median age of 90 years and mild, moderate, or severe frailty scores (n = 3, 7, or 5 respectively). Over the study time course, anti-spike antibody titers were 10-fold higher in individuals with lower frailty status (p = 0.001 and p = 0.005, unadjusted and adjusted for prior COVID-19 infection). Following the booster, titers to spike protein improved regardless of COVID-19 infection or degree of frailty (p = 0.82 and p = 0.29, respectively). Antibody avidity significantly declined over 6 months in all participants following 2 vaccine doses (p < 0.001), which was further impaired with higher frailty (p = 0.001). Notably, avidity increased to peak levels after the booster (p < 0.001). Overall antibody response was inversely correlated with a phenotype of immune-senescent T cells, CD8 + CD28- TEMRA cells (p = 0.036, adjusted for COVID-19 infection). Furthermore, there was increased detection of CD8 + CD28- TEMRA cells in individuals with greater frailty (p = 0.056, adjusted for COVID-19). CONCLUSIONS: We evaluated the immune responses to the Moderna COVID-19 mRNA vaccine in frail older adults in a retirement community. A higher degree of frailty was associated with diminished antibody quantity and quality. However, a booster vaccine dose at 6 months overcame these effects. Frailty was associated with an increased immune-senescence phenotype that may contribute to the observed changes in the vaccine response. While the strength of our conclusions was limited by a small cohort, these results are important for guiding further investigation of vaccine responses in frail older adults.

2.
J Immunol ; 205(8): 2128-2136, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32878911

ABSTRACT

T regulatory cells (Tregs) play a critical role in controlling the immune response, often limiting pathogen-specific cells to curb immune-mediated damage. Studies in human infants have reported an increased representation of Tregs in these individuals. However, how these cells differ from those in adults at various sites and how they respond to activation signals is relatively unknown. In this study, we used a newborn nonhuman primate model to assess Treg populations present at multiple sites with regard to frequency and phenotype in comparison with those present in adult animals. We found that Foxp3+ cells were more highly represented in the T cell compartment of newborn nonhuman primates for all sites examined (i.e., the spleen, lung, and circulation). In the spleen and circulation, newborn-derived Tregs expressed significantly higher levels of Foxp3 and CD25 compared with adults, consistent with an effector phenotype. Strikingly, the phenotype of Tregs in the lungs of adult and infant animals was relatively similar, with both adult and newborn Tregs exhibiting a more uniform PD-1+CD39+ phenotype. Finally, in vitro, newborn Tregs exhibited an increased requirement for TCR engagement for survival. Further, these cells upregulated CD39 more robustly than their adult counterpart. Together, these data provide new insights into the quantity of Tregs in newborns, their activation state, and their potential to respond to activation signals.


Subject(s)
Aging/immunology , Antigens, CD/immunology , Apyrase/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/immunology , Up-Regulation/immunology , Animals , Animals, Newborn , Chlorocebus aethiops , Organ Specificity/immunology , T-Lymphocytes, Regulatory/cytology
3.
J Infect Dis ; 224(2): 351-359, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33245745

ABSTRACT

Eliciting broadly protective antibodies is a critical goal for the development of more effective vaccines against influenza. Optimizing protection is of particular importance in newborns, who are highly vulnerable to severe disease following infection. An effective vaccination strategy for this population must surmount the challenges associated with the neonatal immune system as well as mitigate the inherent immune subdominance of conserved influenza virus epitopes, responses to which can provide broader protection. Here, we show that prime-boost vaccination with a TLR7/8 agonist (R848)-conjugated influenza A virus vaccine elicits antibody responses to the highly conserved hemagglutinin stem and promotes rapid induction of virus neutralizing stem-specific antibodies following viral challenge. These findings support the efficacy of R848 as an effective adjuvant for newborns and demonstrate its ability to enhance antibody responses to subdominant antigenic sites in this at-risk population.


Subject(s)
Antibody Formation , Influenza Vaccines , Orthomyxoviridae Infections , Adjuvants, Immunologic , Animals , Animals, Newborn , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunization, Secondary , Immunoglobulin G/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Primates
4.
Immunology ; 153(3): 357-367, 2018 03.
Article in English | MEDLINE | ID: mdl-28940186

ABSTRACT

Impaired immune responsiveness is a significant barrier to vaccination of neonates. By way of example, the low seroconversion observed following influenza vaccination has led to restriction of its use to infants over 6 months of age, leaving younger infants vulnerable to infection. Our previous studies using a non-human primate neonate model demonstrated that the immune response elicited following vaccination with inactivated influenza virus could be robustly increased by inclusion of the Toll-like receptor agonist flagellin or R848, either delivered individually or in combination. When delivered individually, R848 was found to be the more effective of the two. To gain insights into the mechanism through which these adjuvants functioned in vivo, we assessed the initiation of the immune response, i.e. at 24 hr, in the draining lymph node of neonate non-human primates. Significant up-regulation of co-stimulatory molecules on dendritic cells could be detected, but only when both adjuvants were present. In contrast, R848 alone could increase the number of cells in the lymph node, presumably through enhanced recruitment, as well as B-cell activation at this early time-point. These changes were not observed with flagellin and the dual adjuvanted vaccine did not promote increases beyond those observed with R848 alone. In vitro studies showed that R848 could promote B-cell activation, supporting a model wherein a direct effect on neonate B-cell activation is an important component of the in vivo potency of R848 in neonates.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Animals, Newborn/immunology , B-Lymphocytes/immunology , Imidazoles/immunology , Influenza Vaccines/immunology , Lymph Nodes/immunology , Animals , Antibodies, Viral/immunology , Chlorocebus aethiops , Dendritic Cells/immunology , Flagellin/immunology , Lymphocyte Activation/immunology , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Primates , Vaccination/methods
5.
J Immunol ; 197(2): 555-64, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27279374

ABSTRACT

Influenza virus infection of neonates poses a major health concern, often resulting in severe disease and hospitalization. At present, vaccines for this at-risk population are lacking. Thus, development of an effective vaccine is an urgent need. In this study, we have used an innovative nonhuman primate neonate challenge model to test the efficacy of a novel TLR 7/8 agonist R848-conjugated influenza virus vaccine. The use of the intact virus represents a step forward in conjugate vaccine design because it provides multiple antigenic targets allowing for elicitation of a broad immune response. Our results show that this vaccine induces high-level virus-specific Ab- and cell-mediated responses in neonates that result in increased virus clearance and reduced lung pathology postchallenge compared with the nonadjuvanted virus vaccine. Surprisingly, the addition of a second TLR agonist (flagellin) did not enhance vaccine protection, suggesting that combinations of TLR that provide increased efficacy must be determined empirically. These data support further exploration of this new conjugate influenza vaccine approach as a platform for use in the at-risk neonate population.


Subject(s)
Imidazoles/administration & dosage , Influenza Vaccines/immunology , Vaccines, Inactivated/immunology , Animals , Animals, Newborn , Antibodies, Viral/analysis , Chlorocebus aethiops , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Flagellin/administration & dosage , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism
6.
J Lipid Res ; 58(1): 236-246, 2017 01.
Article in English | MEDLINE | ID: mdl-27811230

ABSTRACT

G protein-coupled receptor (GPR)120/FFA receptor (FFAR)4 (GPR120/FFAR4) activation by n-3 PUFAs attenuates inflammation, but its impact on atherosclerosis is unknown. We determined whether in vivo activation of leukocyte GPR120/FFAR4 by n-3 versus n-6 PUFAs is atheroprotective. Leukocyte GPR120/FFAR4 WT or KO mice in the LDL receptor KO background were generated by bone marrow transplantation. Mice were fed one of the four atherogenic diets containing 0.2% cholesterol and 10% calories as palm oil (PO) + 10% calories as: 1) PO, 2) fish oil (FO; 20:5 n-3 and 22:6 n-3 enriched), 3) echium oil (EO; 18:4 n-3 enriched), or 4) borage oil (BO; 18:3 n-6 enriched) for 16 weeks. Compared with PO, mice fed BO, EO, and FO had significantly reduced plasma cholesterol, TG, VLDL cholesterol, hepatic neutral lipid, and atherosclerosis that were equivalent for WT and KO mice. In BO-, EO-, and FO-fed mice, but not PO-fed mice, lack of leukocyte GPR120/FFAR4 resulted in neutrophilia, pro-inflammatory Ly6Chi monocytosis, increased aortic root monocyte recruitment, and increased hepatic inflammatory gene expression. In conclusion, leukocyte GPR120 expression has minimal effects on dietary PUFA-induced plasma lipid/lipoprotein reduction and atheroprotection, and there is no distinction between n-3 versus n-6 PUFAs in activating anti-inflammatory effects of leukocyte GPR120/FFAR4 in vivo.


Subject(s)
Atherosclerosis/genetics , Leukocytes/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, LDL/genetics , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol/metabolism , Diet, Atherogenic , Fatty Acids, Omega-3/genetics , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/genetics , Fatty Acids, Omega-6/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Leukocytes/pathology , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/metabolism , Transcriptional Activation/genetics
7.
Infect Immun ; 85(4)2017 04.
Article in English | MEDLINE | ID: mdl-28096183

ABSTRACT

Even in the vaccine era, Streptococcus pneumoniae (the pneumococcus) remains a leading cause of otitis media, a significant public health burden, in large part because of the high prevalence of nasal colonization with the pneumococcus in children. The primary pneumococcal neuraminidase, NanA, which is a sialidase that catalyzes the cleavage of terminal sialic acids from host glycoconjugates, is involved in both of these processes. Coinfection with influenza A virus, which also expresses a neuraminidase, exacerbates nasal colonization and disease by S. pneumoniae, in part via the synergistic contributions of the viral neuraminidase. The specific role of its pneumococcal counterpart, NanA, in this interaction, however, is less well understood. We demonstrate in a mouse model that NanA-deficient pneumococci are impaired in their ability to cause both nasal colonization and middle ear infection. Coinfection with neuraminidase-expressing influenza virus and S. pneumoniae potentiates both colonization and infection but not to wild-type levels, suggesting an intrinsic role of NanA. Using in vitro models, we show that while NanA contributes to both epithelial adherence and biofilm viability, its effect on the latter is actually independent of its sialidase activity. These data indicate that NanA contributes both enzymatically and nonenzymatically to pneumococcal pathogenesis and, as such, suggest that it is not a redundant bystander during coinfection with influenza A virus. Rather, its expression is required for the full synergism between these two pathogens.


Subject(s)
Biofilms , Influenza A virus/physiology , Neuraminidase/metabolism , Otitis Media/microbiology , Otitis Media/virology , Streptococcus pneumoniae/physiology , Symbiosis , Animals , Bacterial Adhesion , Disease Models, Animal , Enzyme Activation , Female , Mice , Nasal Mucosa/microbiology , Neuraminidase/genetics
8.
J Virol ; 89(14): 7291-303, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25948746

ABSTRACT

UNLABELLED: Influenza virus can cause life-threatening infections in neonates and young infants. Although vaccination is a major countermeasure against influenza, current vaccines are not approved for use in infants less than 6 months of age, in part due to the weak immune response following vaccination. Thus, there is a strong need to develop new vaccines with improved efficacy for this vulnerable population. To address this issue, we established a neonatal African green monkey (AGM) nonhuman primate model that could be used to identify effective influenza vaccine approaches for use in young infants. We assessed the ability of flagellin, a Toll-like receptor 5 (TLR5) agonist, to serve as an effective adjuvant in this at-risk population. Four- to 6-day-old AGMs were primed and boosted with inactivated PR8 influenza virus (IPR8) adjuvanted with either wild-type flagellin or inactive flagellin with a mutation at position 229 (m229), the latter of which is incapable of signaling through TLR5. Increased IgG responses were observed following a boost, as well as at early times after challenge, in infants vaccinated with flagellin-adjuvanted IPR8. Inclusion of flagellin during vaccination also resulted in a significantly increased number of influenza virus-specific T cells following challenge compared to the number in infants vaccinated with the m229 adjuvant. Finally, following challenge infants vaccinated with IPR8 plus flagellin exhibited a reduced pathology in the lungs compared to that in infants that received IPR8 plus m229. This study provides the first evidence of flagellin-mediated enhancement of vaccine responses in nonhuman primate neonates. IMPORTANCE: Young infants are particularly susceptible to severe disease as a result of influenza virus infection. Compounding this is the lack of effective vaccines for use in this vulnerable population. Here we describe a vaccine approach that results in improved immune responses and protection in young infants. Incorporation of flagellin during vaccination resulted in increased antibody and T cell responses together with reduced disease following virus infection. These results suggest that flagellin may serve as an effective adjuvant for vaccines targeted to this vulnerable population.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Flagellin/administration & dosage , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Vaccination/methods , Animals , Animals, Newborn , Antibodies, Viral/blood , Chlorocebus aethiops , Disease Models, Animal , Immunoglobulin G/blood , Influenza Vaccines/administration & dosage , T-Lymphocytes/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
9.
J Immunol ; 193(11): 5363-9, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25411431

ABSTRACT

The first six months of life reflect a time of high susceptibility to severe disease following respiratory virus infection. Although this could be improved significantly by immunization, current vaccines are not approved for use in these very young individuals. This is the result of the combined effects of poor immune responsiveness and safety concerns regarding the use of live attenuated vaccines or potent adjuvants in this population. Vaccines to effectively combat respiratory viral infection ideally would result in robust CD4(+) and CD8(+) T cell responses, as well as high-affinity Ab. Inclusion of TLR agonists or single-cycle viruses is an attractive approach for provision of signals that can act as potent stimulators of dendritic cell maturation, as well as direct activators of T and/or B cells. In this article, I discuss the challenges associated with generation of a robust immune response in neonates and the potential for adjuvants to overcome these obstacles.


Subject(s)
Respiratory Tract Infections/immunology , Viral Vaccines , Virus Diseases/immunology , Viruses/immunology , Adjuvants, Immunologic , Animals , Antibodies, Viral/immunology , Humans , Infant, Newborn , Respiratory Tract Infections/prevention & control , Vaccination , Virus Diseases/prevention & control
10.
J Immunol ; 193(10): 5076-87, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25311807

ABSTRACT

Infection with influenza A virus can lead to increased susceptibility to subsequent bacterial infection, often with Streptococcus pneumoniae. Given the substantial modification of the lung environment that occurs following pathogen infection, there is significant potential for modulation of immune responses. In this study, we show that infection of mice with influenza virus, followed by the noninvasive EF3030 strain of Streptococcus pneumoniae, leads to a significant decrease in the virus-specific CD8(+) T cell response in the lung. Adoptive-transfer studies suggest that this reduction contributes to disease in coinfected animals. The reduced number of lung effector cells in coinfected animals was associated with increased death, as well as a reduction in cytokine production in surviving cells. Further, cells that retained the ability to produce IFN-γ exhibited a decreased potential for coproduction of TNF-α. Reduced cytokine production was directly correlated with a decrease in the level of mRNA. Negative regulation of cells in the mediastinal lymph node was minimal compared with that present in the lung, supporting a model of selective regulation in the tissue harboring high pathogen burden. These results show that entry of a coinfecting pathogen can have profound immunoregulatory effects on an ongoing immune response. Together, these findings reveal a novel dynamic interplay between concurrently infecting pathogens and the adaptive immune system.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Lung/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Pneumococcal/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , Bacterial Load , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Coinfection , Female , Immunomodulation , Influenza A Virus, H1N1 Subtype/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Lung/microbiology , Lung/pathology , Lung/virology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Lymph Nodes/virology , Mice , Mice, Inbred BALB C , Organ Specificity , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology , Severity of Illness Index , Streptococcus pneumoniae/immunology , Survival Analysis , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Viral Load
11.
J Immunol ; 190(10): 5020-9, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23589620

ABSTRACT

Generating and maintaining a robust CD8(+) T cell response in the face of high viral burden is vital for host survival. Further, balancing the differentiation of effectors along the memory precursor effector cell pathway versus the short-lived effector cell (SLEC) pathway may be critical in controlling the outcome of virus infection with regard to clearance and establishing protection. Although recent studies have identified several factors that have the capacity to regulate effector CD8(+) T cell differentiation-for example, inflammatory cytokines-we are far from a complete understanding of how cells choose the memory precursor effector cell versus SLEC fate following infection. In this study, we have modulated the infectious dose of the poxvirus vaccinia virus as an approach to modulate the environment present during activation and expansion of virus-specific effector cells. Surprisingly, in the face of a high virus burden, the number of SLECs was decreased. This decrease was the result of increased natural regulatory T cells (Tregs) generated by high viral burden, as depletion of these cells restored SLECs. Our data suggest Treg modulation of differentiation occurs via competition for IL-2 during the late expansion period, as opposed to the time of T cell priming. These findings support a novel model wherein modulation of the Treg response as a result of high viral burden regulates late-stage SLEC number.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/virology , CD8-Positive T-Lymphocytes/virology , Vaccinia virus/immunology , Vaccinia/immunology , Animals , B-Lymphocyte Subsets/metabolism , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/metabolism , Caspase 3/metabolism , Cell Differentiation/immunology , Cell Proliferation , Female , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Interleukin-12/metabolism , Interleukin-2 , Interleukin-2 Receptor alpha Subunit/metabolism , Lectins, C-Type , Mice , Mice, Inbred C57BL , Receptors, Immunologic , STAT5 Transcription Factor , T-Lymphocytes, Regulatory/virology , Viral Load
12.
Infect Immun ; 82(11): 4802-12, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25156728

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is both a widespread nasal colonizer and a leading cause of otitis media, one of the most common diseases of childhood. Pneumococcal phase variation influences both colonization and disease and thus has been linked to the bacteria's transition from colonizer to otopathogen. Further contributing to this transition, coinfection with influenza A virus has been strongly associated epidemiologically with the dissemination of pneumococci from the nasopharynx to the middle ear. Using a mouse infection model, we demonstrated that coinfection with influenza virus and pneumococci enhanced both colonization and inflammatory responses within the nasopharynx and middle ear chamber. Coinfection studies were also performed using pneumococcal populations enriched for opaque or transparent phase variants. As shown previously, opaque variants were less able to colonize the nasopharynx. In vitro, this phase also demonstrated diminished biofilm viability and epithelial adherence. However, coinfection with influenza virus ameliorated this colonization defect in vivo. Further, viral coinfection ultimately induced a similar magnitude of middle ear infection by both phase variants. These data indicate that despite inherent differences in colonization, the influenza A virus exacerbation of experimental middle ear infection is independent of the pneumococcal phase. These findings provide new insights into the synergistic link between pneumococcus and influenza virus in the context of otitis media.


Subject(s)
Influenza A virus , Nose/microbiology , Orthomyxoviridae Infections/complications , Otitis Media/microbiology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/physiology , Animals , Carrier State , Coinfection , Mice , Otitis Media/complications , Pneumococcal Infections/complications
13.
J Immunol ; 189(2): 638-45, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22706075

ABSTRACT

By the peak of the CD8(+) T cell response, the effector cell pool consists of a heterogeneous population of cells that includes both those with an increased propensity to become long-lived memory cells (memory precursor effector cells; MPEC) and those that are terminally differentiated cells (short-lived effector cells; SLEC). Numerous studies have established the critical role that functional avidity plays in determining the in vivo efficacy of CD8(+) effector cells. Currently, how functional avidity differs in MPEC versus SLEC and the evolution of this property within these two populations during the expansion and contraction of the response are unknown. The data presented in this study show that at the peak of the effector response generated after poxvirus infection, SLEC were of higher functional avidity than their MPEC counterpart. Over time, however, SLEC exhibited a decrease in peptide sensitivity. This is in contrast to MPEC, which showed a modest increase in peptide sensitivity as the response reached equilibrium. The decrease in functional avidity in SLEC was independent of CD8 modulation or the amount of Ag receptor expressed by the T cell. Instead, the loss in sensitivity was correlated with decreased expression and activation of ZAP70 and Lck, critical components of TCR membrane proximal signaling. These results highlight the potential contribution of avidity in the differentiation and evolution of the T cell effector response after viral infection.


Subject(s)
CD8 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/immunology , Cellular Senescence/immunology , Immunologic Memory , Lymphocytic choriomeningitis virus/immunology , Vaccinia virus/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , CD8 Antigens/physiology , CD8-Positive T-Lymphocytes/metabolism , Cell Adhesion/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Time Factors
14.
Vaccines (Basel) ; 12(4)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38675797

ABSTRACT

A number of studies have demonstrated the role of sex in regulating immune responses to vaccination. However, these findings have been limited to adults for both human and animal models. As a result, our understanding of the impact of sex on vaccine responses in the newborn is highly limited. Here, we probe this important question using a newborn non-human primate model. We leveraged our prior analysis of two cohorts of newborns, with one being mother-reared and one nursery-reared. This provided adequate numbers of males and females to interrogate the impact of sex on the response to inactivated influenza vaccines alone or adjuvanted with R848, flagellin, or both. We found that, in contrast to what has been reported in adults, the non-adjuvanted inactivated influenza virus vaccine induced similar levels of virus-specific IgG in male and female newborns. However, the inclusion of R848, either alone or in combination with flagellin, resulted in higher antibody titers in females compared to males. Sex-specific increases in the neutralizing antibody were only observed when both R848 and flagellin were present. These data, generated in the highly translational NHP newborn model, provide novel insights into the role of sex in the immune response of newborns.

15.
J Virol ; 86(19): 10640-50, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22811544

ABSTRACT

An increasing number of studies suggest that individual subsets of dendritic cells (DC) exhibit distinct capabilities with regard to the generation of the adaptive immune response. In this study, we evaluated the properties of a relatively unexplored DC subset present in the lung-draining mediastinal lymph node. This subset expresses the airway dendritic cell marker CD103 together with CD8. These DC were of interest given that our previous studies using a model of respiratory infection with vaccinia virus revealed a distinct difference in the ability of CD103(+) DC to prime T cells that correlated inversely with the expression of CD8, suggesting a differential role of these DC in the context of respiratory virus infection. To expand our understanding of the role of this DC population, we performed analyses to elucidate the phenotype, migratory capacity, responsiveness to innate stimuli, and priming capacity of CD8(+) CD103(+) DC. We found that expression of surface markers on these DC was similar to that of CD8(-) CD103(+) DC, supporting their close relationship. Further, the two DC types were similar with regard to antigen uptake. However, although both CD103(+) subsets originated from the lung, CD8-bearing CD103(+) DC appeared in the lymph node with delayed kinetics following virus infection. While this subset exhibited increased responsiveness to a number of Toll-like receptor (TLR) agonists, their response to infection was virus specific, demonstrating poor responsiveness to vaccinia virus infection but robust maturation following infection with parainfluenza virus 5 or influenza virus. These findings show that CD8 marks a population of lung airway-derived DC with distinct migratory and maturation responses that likely contribute differentially to the immune response depending on the infecting pathogen.


Subject(s)
CD8 Antigens/biosynthesis , Dendritic Cells/cytology , Lung/metabolism , Toll-Like Receptors/metabolism , Virus Diseases/metabolism , Animals , Antigens, CD/biosynthesis , Cell Movement , Flow Cytometry/methods , Integrin alpha Chains/biosynthesis , Kinetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Pinocytosis , Vaccinia/metabolism , Vaccinia/virology , Vaccinia virus/metabolism
16.
J Virol ; 86(18): 9929-40, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22761385

ABSTRACT

Enveloped viruses can incorporate host cell membrane proteins during the budding process. Here we demonstrate that mumps virus (MuV) and vesicular stomatitis virus (VSV) assemble to include CD46 and CD55, two host cell regulators which inhibit propagation of complement pathways through distinct mechanisms. Using viruses which incorporated CD46 alone, CD55 alone, or both CD46 and CD55, we have tested the relative contribution of these regulators in resistance to complement-mediated neutralization. Virion-associated CD46 and CD55 were biologically active, with VSV showing higher levels of activity of both cofactors, which promoted factor I-mediated cleavage of C3b into iC3b as well as decay-accelerating factor (DAF) activity against the C3 convertase, than MuV. Time courses of in vitro neutralization with normal human serum (NHS) showed that both regulators could delay neutralization, but viruses containing CD46 alone were neutralized faster and more completely than viruses containing CD55 alone. A dominant inhibitory role for CD55 was most evident for VSV, where virus containing CD55 alone was not substantially different in neutralization kinetics from virus harboring both regulators. Electron microscopy showed that VSV neutralization proceeded through virion aggregation followed by lysis, with virion-associated CD55 providing a delay in both aggregation and lysis more substantial than that conferred by CD46. Our results demonstrate the functional significance of incorporation of host cell factors during virion envelope assembly. They also define pathways of virus complement-mediated neutralization and suggest the design of more effective viral vectors.


Subject(s)
CD55 Antigens/physiology , Complement Activation/physiology , Membrane Cofactor Protein/physiology , Mumps virus/immunology , Vesiculovirus/immunology , Animals , CD55 Antigens/genetics , CHO Cells , Complement Activation/genetics , Cricetinae , Cricetulus , Host-Pathogen Interactions/immunology , Humans , Membrane Cofactor Protein/genetics , Microscopy, Immunoelectron , Mumps virus/physiology , Mumps virus/ultrastructure , Neutralization Tests , Vesiculovirus/physiology , Vesiculovirus/ultrastructure , Virus Assembly
17.
Arterioscler Thromb Vasc Biol ; 32(9): 2122-30, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22814747

ABSTRACT

OBJECTIVE: Fish oil, containing omega-3 fatty acids, attenuates atherosclerosis. We hypothesized that omega-3 fatty acid-enriched oils are atheroprotective through alteration of monocyte subsets and their trafficking into atherosclerotic lesions. METHODS AND RESULTS: Low-density lipoprotein receptor knockout and apolipoprotein E(-/-) mice were fed diets containing 10% (calories) palm oil and 0.2% cholesterol, supplemented with an additional 10% palm oil, echium oil (containing 18:4 n-3), or fish oil. Compared with palm oil-fed low-density lipoprotein receptor knockout mice, echium oil and fish oil significantly reduced plasma cholesterol, splenic Ly6C(hi) monocytosis by ≈50%, atherosclerosis by 40% to 70%, monocyte trafficking into the aortic root by ≈50%, and atherosclerotic lesion macrophage content by 30% to 44%. In contrast, atherosclerosis and monocyte trafficking into the artery wall was not altered by omega-3 fatty acids in apolipoprotein E(-/-) mice; however, Ly6C(hi) splenic monocytes positively correlated with aortic root intimal area across all diet groups. In apolipoprotein E(-/-) mice, fish oil reduced the percentage of blood Ly6C(hi) monocytes, despite an average 2-fold higher plasma cholesterol relative to palm oil. CONCLUSIONS: The presence of splenic Ly6C(hi) monocytes parallels the appearance of atherosclerotic disease in both low-density lipoprotein receptor knockout and apolipoprotein E(-/-) mice. Furthermore, omega-3 fatty acids favorably alter monocyte subsets independently from effects on plasma cholesterol and reduce monocyte recruitment into atherosclerotic lesions.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Chemotaxis/drug effects , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Monocytes/drug effects , Plant Oils/pharmacology , Spleen/drug effects , Animals , Antigens, Ly/blood , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Biomarkers/blood , Cholesterol, Dietary/blood , Disease Models, Animal , Echium , Female , Inflammation Mediators/blood , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Neutrophils/drug effects , Neutrophils/immunology , Palm Oil , Receptors, LDL/deficiency , Receptors, LDL/genetics , Spleen/immunology , Spleen/metabolism , Time Factors
18.
Int J Gynaecol Obstet ; 162(1): 163-168, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36690442

ABSTRACT

OBJECTIVE: The objective of this study was to evaluate hemagglutinin stem-specific antibody response to the influenza vaccine during pregnancy and its transfer to the infant. METHODS: The authors assessed antibody titers among maternal participants and their paired neonate's cord blood (CB) using enzyme-linked immunoassay. Fifteen pregnant participants pre-2019 and post-2019 seasonal influenza vaccine were compared with 18 prenatally vaccinated participants with paired neonatal CB samples. Total IgG and IgG subclass titers specific for whole vaccine antigens versus recombinant hemagglutinin stem-specific antigen were compared using Wilcoxon exact test. RESULTS: Hemagglutinin stem-specific IgG was boosted more robustly than whole vaccine titers when comparing postvaccine versus prevaccine log2 IgG ratios (P = 0.04). Hemagglutinin stem-specific IgG titers were boosted postvaccination (prevaccine: 14.5 [95% confidence interval, 13.8-15.2] vs. postvaccine: 16 [95% confidence interval, 15.2-16.8], P = 0.004). While IgG to whole vaccine was similar in neonatal CB and maternal plasma (P = 0.09), hemagglutinin stem-specific IgG concentrated in CB (P = 0.002), which was dominated by IgG1 subclass (analysis of variance P < 0.05). CONCLUSION: These data demonstrate the ability of pregnant women to generate a more robust antibody response to the stem region compared with the head region of hemagglutinin with transplacental transfer of IgG.


Subject(s)
Influenza Vaccines , Influenza, Human , Infant , Infant, Newborn , Humans , Female , Pregnancy , Hemagglutinins , Influenza, Human/prevention & control , Vaccination , Immunoglobulin G , Antibodies, Viral
19.
Vaccines (Basel) ; 11(7)2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37515076

ABSTRACT

Subunit or inactivated vaccines comprise the majority of vaccines used against viral and bacterial pathogens. However, compared to their live/attenuated counterparts, these vaccines often demonstrate reduced immunogenicity, requiring multiple boosters and or adjuvants to elicit protective immune responses. For this reason, studies of adjuvants and the mechanism through which they can improve inactivated vaccine responses are critical for the development of vaccines with increased efficacy. Studies have shown that the direct conjugation of adjuvant to antigen promotes vaccine immunogenicity, with the advantage of both the adjuvant and antigen targeting the same cell. Using this strategy of direct linkage, we developed an inactivated influenza A (IAV) vaccine that is directly conjugated with the Toll-like receptor 7/8 agonist resiquimod (R848) through a heterobifunctional crosslinker. Previously, we showed that this vaccine resulted in improved protection and viral clearance in newborn nonhuman primates compared to a non-adjuvanted vaccine. We subsequently discovered that the choice of linker used to conjugate R848 to the virus alters the stimulatory activity of the vaccine, promoting increased maturation and proinflammatory cytokine production from DC differentiated in vitro. With this knowledge, we explored how the choice of crosslinker impacts the stimulatory activity of these vaccines. We found that the linker choice alters signaling through the NF-κB pathway in human monocyte-derived dendritic cells (moDCs). Further, we extended our analyses to in vivo differentiated APC present in human peripheral blood, replicating the linker-dependent differences found in in vitro differentiated cells. Finally, we demonstrated in a mouse model that the choice of linker impacts the amount of IAV-specific IgG antibody produced in response to vaccination. These data enhance our understanding of conjugation approaches for improving vaccine immunogenicity.

20.
Vaccines (Basel) ; 10(4)2022 Mar 23.
Article in English | MEDLINE | ID: mdl-35455242

ABSTRACT

Elderly individuals are highly susceptible to developing severe outcomes as a result of influenza A virus (IAV) infection. This can be attributed to alterations that span the aged immune system, which also result in reduced responsiveness to the seasonal inactivated vaccine. Given the rapidly increasing number of individuals in this age group, it is imperative that we develop strategies that can better protect this population from IAV-associated disease. Based on our previous findings that the TLR7/8 agonist resiquimod (R848) could efficiently boost responses in the newborn, another population with decreased vaccine responsiveness, we evaluated this adjuvant in an elderly African green monkey (AGM) model. AGM aged 16-24 years old (equivalent to 64-96 in human years) were primed and boosted with inactivated A/PuertoRico/8/1934 (H1N1) (IPR8) alone or directly linked to R848 (IPR8-R848). We observed increases in the level of circulating virus-specific IgM antibody 10 days following primary vaccination in AGM that were vaccinated with IPR8-R848, but not IPR8 alone. In addition, there were significant increases in virus-specific IgG after boosting selectively in the IPR8-R848 vaccinated animals. These findings provide insights into the ability of R848 to modulate the aged immune system in the context of IAV vaccination.

SELECTION OF CITATIONS
SEARCH DETAIL