Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32759316

ABSTRACT

An entirely plasmid-based reverse genetics (RG) system was recently developed for rotavirus (RV), opening new avenues for in-depth molecular dissection of RV biology, immunology, and pathogenesis. Several improvements to further optimize the RG efficiency have now been described. However, only a small number of individual RV strains have been recovered to date. None of the current methods have supported the recovery of murine RV, impeding the study of RV replication and pathogenesis in an in vivo suckling mouse model. Here, we describe useful modifications to the RG system that significantly improve rescue efficiency of multiple RV strains. In addition to the 11 group A RV segment-specific (+)RNAs [(+)ssRNAs], a chimeric plasmid was transfected, from which the capping enzyme NP868R of African swine fever virus (ASFV) and the T7 RNA polymerase were expressed. Second, a genetically modified MA104 cell line was used in which several components of the innate immunity were degraded. Using this RG system, we successfully recovered the simian RV RRV strain, the human RV CDC-9 strain, a reassortant between murine RV D6/2 and simian RV SA11 strains, and several reassortants and reporter RVs. All these recombinant RVs were rescued at a high efficiency (≥80% success rate) and could not be reliably rescued using several recently published RG strategies (<20%). This improved system represents an important tool and great potential for the rescue of other hard-to-recover RV strains such as low-replicating attenuated vaccine candidates or low-cell culture passage clinical isolates from humans or animals.IMPORTANCE Group A rotavirus (RV) remains as the single most important cause of severe acute gastroenteritis among infants and young children worldwide. An entirely plasmid-based reverse genetics (RG) system was recently developed, opening new ways for in-depth molecular study of RV. Despite several improvements to further optimize the RG efficiency, it has been reported that current strategies do not enable the rescue of all cultivatable RV strains. Here, we described a helpful modification to the current strategies and established a tractable RG system for the rescue of the simian RRV strain, the human CDC-9 strain, and a murine-like RV strain, which is suitable for both in vitro and in vivo studies. This improved RV reverse genetics system will facilitate study of RV biology in both in vitro and in vivo systems that will facilitate the improved design of RV vaccines, better antiviral therapies, and expression vectors.


Subject(s)
Gene Expression Regulation, Viral , Host-Pathogen Interactions/genetics , Reassortant Viruses/genetics , Reverse Genetics/methods , Rotavirus/genetics , Viral Proteins/genetics , African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , Animals , Chlorocebus aethiops , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Mice , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Plasmids/chemistry , Plasmids/metabolism , RNA Caps , Reassortant Viruses/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Rotavirus/immunology , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Transfection , Vero Cells , Viral Proteins/immunology , Virus Replication
2.
Gastroenterology ; 159(1): 214-226.e1, 2020 07.
Article in English | MEDLINE | ID: mdl-32247021

ABSTRACT

BACKGROUND & AIMS: Intestinal microfold (M) cells are a unique subset of intestinal epithelial cells in the Peyer's patches that regulate mucosal immunity, serving as portals for sampling and uptake of luminal antigens. The inability to efficiently develop human M cells in cell culture has impeded studies of the intestinal immune system. We aimed to identify signaling pathways required for differentiation of human M cells and establish a robust culture system using human ileum enteroids. METHODS: We analyzed transcriptome data from mouse Peyer's patches to identify cell populations in close proximity to M cells. We used the human enteroid system to determine which cytokines were required to induce M-cell differentiation. We performed transcriptome, immunofluorescence, scanning electron microscope, and transcytosis experiments to validate the development of phenotypic and functional human M cells. RESULTS: A combination of retinoic acid and lymphotoxin induced differentiation of glycoprotein 2-positive human M cells, which lack apical microvilli structure. Upregulated expression of innate immune-related genes within M cells correlated with a lack of viral antigens after rotavirus infection. Human M cells, developed in the enteroid system, internalized and transported enteric viruses, such as rotavirus and reovirus, across the intestinal epithelium barrier in the enteroids. CONCLUSIONS: We identified signaling pathways required for differentiation of intestinal M cells, and used this information to create a robust culture method to develop human M cells with capacity for internalization and transport of viruses. Studies of this model might increase our understanding of antigen presentation and the systemic entry of enteric pathogens in the human intestine.


Subject(s)
Cell Differentiation/immunology , Lymphotoxin-alpha/metabolism , Peyer's Patches/immunology , Signal Transduction/immunology , Tretinoin/metabolism , Animals , Antigen Presentation/immunology , Cell Culture Techniques/methods , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Ileum/cytology , Ileum/immunology , Immunity, Mucosal , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Mice , NF-kappa B/metabolism , Organoids , Peyer's Patches/cytology , Peyer's Patches/metabolism , Primary Cell Culture , Recombinant Proteins/metabolism
3.
J Virol ; 94(9)2020 04 16.
Article in English | MEDLINE | ID: mdl-32051268

ABSTRACT

Our understanding of how rotavirus (RV) subverts host innate immune signaling has greatly increased over the past decade. However, the relative contribution of each virus-encoded innate immune antagonist has not been fully studied in the context of RV infection in vivo Here, we present both in vitro and in vivo evidence that the host interferon (IFN)-inducible 2'-5'-oligoadenylate synthetase (OAS) and RNase L pathway effectively suppresses the replication of heterologous RV strains. VP3 from homologous RVs relies on its 2'-5'-phosphodiesterase (PDE) domain to counteract RNase L-mediated antiviral signaling. Using an RV reverse-genetics system, we show that compared to the parental strain, VP3 PDE mutant RVs replicated at low levels in the small intestine and were shed less in the feces of wild-type mice, and such defects were rescued in Rnasel-/- suckling mice. Collectively, these findings highlight an important role of VP3 in promoting viral replication and pathogenesis in vivo in addition to its well-characterized function as the viral RNA-capping enzyme.IMPORTANCE Rotaviruses are significant human pathogens that result in diarrhea, dehydration, and deaths in many children around the world. Rotavirus vaccines have suboptimal efficacy in low- to middle-income countries, where the burden of the diseases is the most severe. With the ultimate goal of improving current vaccines, we aim to better understand how rotavirus interacts with the host innate immune system in the small intestine. Here, we demonstrate that interferon-activated RNase L signaling blocks rotavirus replication in a strain-specific manner. In addition, virus-encoded VP3 antagonizes RNase L activity both in vitro and in vivo These studies highlight an ever-evolving arms race between antiviral factors and viral pathogens and provide a new means of targeted attenuation for next-generation rotavirus vaccine design.


Subject(s)
Capsid Proteins/genetics , Endoribonucleases/genetics , Rotavirus/genetics , Adenine Nucleotides/metabolism , Animals , Capsid Proteins/metabolism , Cell Line , Chlorocebus aethiops , Endoribonucleases/metabolism , Female , Host-Pathogen Interactions/genetics , Immunity, Innate/immunology , Interferons/metabolism , Male , Mice , Mice, Inbred C57BL , Oligoribonucleotides/metabolism , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Polynucleotide Ligases/metabolism , Reverse Genetics/methods , Rotavirus Infections/virology , Rotavirus Vaccines , Signal Transduction/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/genetics
4.
J Pediatric Infect Dis Soc ; 9(6): 752-755, 2020 Dec 31.
Article in English | MEDLINE | ID: mdl-31644795

ABSTRACT

In this study, we illustrate, for the first time, that preexisting low-avidity neutralizing measles maternal antibodies do not interfere with the development of high concentrations of high-avidity measles antibodies in children immunized at age 12 months. This suggests that the quality of measles maternal antibodies, rather than the quantity, impacts immunogenicity of primary measles immunization.


Subject(s)
Measles , Mumps , Antibodies, Viral , Antibody Formation , Child , Humans , Infant , Measles/prevention & control , Measles Vaccine , Measles-Mumps-Rubella Vaccine
5.
J Clin Invest ; 129(9): 3839-3851, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31403468

ABSTRACT

We previously generated 32 rotavirus-specific (RV-specific) recombinant monoclonal antibodies (mAbs) derived from B cells isolated from human intestinal resections. Twenty-four of these mAbs were specific for the VP8* fragment of RV VP4, and most (20 of 24) were non-neutralizing when tested in the conventional MA104 cell-based assay. We reexamined the ability of these mAbs to neutralize RVs in human intestinal epithelial cells including ileal enteroids and HT-29 cells. Most (18 of 20) of the "non-neutralizing" VP8* mAbs efficiently neutralized human RV in HT-29 cells or enteroids. Serum RV neutralization titers in adults and infants were significantly higher in HT-29 than MA104 cells and adsorption of these sera with recombinant VP8* lowered the neutralization titers in HT-29 but not MA104 cells. VP8* mAbs also protected suckling mice from diarrhea in an in vivo challenge model. X-ray crystallographic analysis of one VP8* mAb (mAb9) in complex with human RV VP8* revealed that the mAb interaction site was distinct from the human histo-blood group antigen binding site. Since MA104 cells are the most commonly used cell line to detect anti-RV neutralization activity, these findings suggest that prior vaccine and other studies of human RV neutralization responses may have underestimated the contribution of VP8* antibodies to the overall neutralization titer.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Epithelial Cells/immunology , Intestines/cytology , Rotavirus Infections/immunology , Adsorption , Animals , Antigens, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/virology , Binding Sites , Caco-2 Cells , Cell Line , Crystallography, X-Ray , Epithelial Cells/virology , Genotype , Haplorhini , Humans , Immunoglobulin G/chemistry , India , Infant , Infant, Newborn , Intestines/virology , Mice , Neutralization Tests , Polysaccharides/chemistry , Protein Conformation , Recombinant Proteins/immunology , United States
6.
Nat Commun ; 9(1): 1485, 2018 04 16.
Article in English | MEDLINE | ID: mdl-29662124

ABSTRACT

Cohesin is a multi-subunit nuclear protein complex that coordinates sister chromatid separation during cell division. Highly frequent somatic mutations in genes encoding core cohesin subunits have been reported in multiple cancer types. Here, using a genome-wide CRISPR-Cas9 screening approach to identify host dependency factors and novel innate immune regulators of rotavirus (RV) infection, we demonstrate that the loss of STAG2, an important component of the cohesin complex, confers resistance to RV replication in cell culture and human intestinal enteroids. Mechanistically, STAG2 deficiency results in spontaneous genomic DNA damage and robust interferon (IFN) expression via the cGAS-STING cytosolic DNA-sensing pathway. The resultant activation of JAK-STAT signaling and IFN-stimulated gene (ISG) expression broadly protects against virus infections, including RVs. Our work highlights a previously undocumented role of the cohesin complex in regulating IFN homeostasis and identifies new therapeutic avenues for manipulating the innate immunity.


Subject(s)
Antigens, Nuclear/immunology , Cell Cycle Proteins/immunology , Chromosomal Proteins, Non-Histone/immunology , Host-Pathogen Interactions , Membrane Proteins/immunology , Nucleotidyltransferases/immunology , Rotavirus/immunology , Spheroids, Cellular/immunology , Antigens, Nuclear/genetics , CRISPR-Cas Systems , Caco-2 Cells , Cell Cycle Proteins/genetics , Cell Nucleus/immunology , Cell Nucleus/virology , Chromosomal Proteins, Non-Histone/genetics , DNA Damage , Gene Deletion , Gene Editing , Gene Expression Regulation , Genome, Human , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Interferons/genetics , Interferons/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Janus Kinases/genetics , Janus Kinases/immunology , Membrane Proteins/genetics , Nucleotidyltransferases/genetics , Rotavirus/growth & development , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , Signal Transduction , Spheroids, Cellular/virology , Cohesins
7.
Proc Natl Acad Sci U S A ; 114(18): E3642-E3651, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28416666

ABSTRACT

Despite the wide administration of several effective vaccines, rotavirus (RV) remains the single most important etiological agent of severe diarrhea in infants and young children worldwide, with an annual mortality of over 200,000 people. RV attachment and internalization into target cells is mediated by its outer capsid protein VP4. To better understand the molecular details of RV entry, we performed tandem affinity purification coupled with high-resolution mass spectrometry to map the host proteins that interact with VP4. We identified an actin-binding protein, drebrin (DBN1), that coprecipitates and colocalizes with VP4 during RV infection. Importantly, blocking DBN1 function by siRNA silencing, CRISPR knockout (KO), or chemical inhibition significantly increased host cell susceptibility to RV infection. Dbn1 KO mice exhibited higher incidence of diarrhea and more viral antigen shedding in their stool samples compared with the wild-type littermates. In addition, we found that uptake of other dynamin-dependent cargos, including transferrin, cholera toxin, and multiple viruses, was also enhanced in DBN1-deficient cells. Inhibition of cortactin or dynamin-2 abrogated the increased virus entry observed in DBN1-deficient cells, suggesting that DBN1 suppresses dynamin-mediated endocytosis via interaction with cortactin. Our study unveiled an unexpected role of DBN1 in restricting the entry of RV and other viruses into host cells and more broadly to function as a crucial negative regulator of diverse dynamin-dependent endocytic pathways.


Subject(s)
Dynamins/metabolism , Endocytosis , Neuropeptides/metabolism , Rotavirus Infections/metabolism , Rotavirus/metabolism , Virus Internalization , Animals , Cricetinae , Dynamin II , Dynamins/genetics , HEK293 Cells , Humans , Mice , Mice, Knockout , Neuropeptides/genetics , Rotavirus/genetics , Rotavirus Infections/genetics
9.
PLoS Pathog ; 12(4): e1005600, 2016 04.
Article in English | MEDLINE | ID: mdl-27128797

ABSTRACT

Type I (IFN-α/ß) and type III (IFN-λ) interferons (IFNs) exert shared antiviral activities through distinct receptors. However, their relative importance for antiviral protection of different organ systems against specific viruses remains to be fully explored. We used mouse strains deficient in type-specific IFN signaling, STAT1 and Rag2 to dissect distinct and overlapping contributions of type I and type III IFNs to protection against homologous murine (EW-RV strain) and heterologous (non-murine) simian (RRV strain) rotavirus infections in suckling mice. Experiments demonstrated that murine EW-RV is insensitive to the action of both types of IFNs, and that timely viral clearance depends upon adaptive immune responses. In contrast, both type I and type III IFNs can control replication of the heterologous simian RRV in the gastrointestinal (GI) tract, and they cooperate to limit extra-intestinal simian RRV replication. Surprisingly, intestinal epithelial cells were sensitive to both IFN types in neonatal mice, although their responsiveness to type I, but not type III IFNs, diminished in adult mice, revealing an unexpected age-dependent change in specific contribution of type I versus type III IFNs to antiviral defenses in the GI tract. Transcriptional analysis revealed that intestinal antiviral responses to RV are triggered through either type of IFN receptor, and are greatly diminished when receptors for both IFN types are lacking. These results also demonstrate a murine host-specific resistance to IFN-mediated antiviral effects by murine EW-RV, but the retention of host efficacy through the cooperative action by type I and type III IFNs in restricting heterologous simian RRV growth and systemic replication in suckling mice. Collectively, our findings revealed a well-orchestrated spatial and temporal tuning of innate antiviral responses in the intestinal tract where two types of IFNs through distinct patterns of their expression and distinct but overlapping sets of target cells coordinately regulate antiviral defenses against heterologous or homologous rotaviruses with substantially different effectiveness.


Subject(s)
Interferon Type I/immunology , Interferon-gamma/immunology , Intestines/immunology , Rotavirus Infections/immunology , Animals , Animals, Newborn , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Rotavirus
10.
J Virol ; 87(15): 8307-16, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23698306

ABSTRACT

Homologous rotaviruses (RV) are, in general, more virulent and replicate more efficiently than heterologous RV in the intestine of the homologous host. The genetic basis for RV host range restriction is not fully understood and is likely to be multigenic. In previous studies, RV genes encoding VP3, VP4, VP7, nonstructural protein 1 (NSP1), and NSP4 have all been implicated in strain- and host species-specific infection. These studies used different RV strains, variable measurements of host range, and different animal hosts, and no clear consensus on the host range restriction determinants emerged. We used a murine model to demonstrate that enteric replication of murine RV EW is 1,000- to 10,000-fold greater than that of a simian rotavirus (RRV) in suckling mice. Intestinal replication of a series of EW × RRV reassortants was used to identify several RV genes that influenced RV replication in the intestine. The role of VP4 (encoded by gene 4) in enteric infection was strain specific. RRV VP4 reduced murine RV infectivity only slightly; however, a reassortant expressing VP4 from a bovine RV strain (UK) severely restricted intestinal replication in the suckling mice. The homologous murine EW NSP1 (encoded by gene 5) was necessary but not sufficient for promoting efficient enteric growth. Efficient enteric replication required a constellation of murine genes encoding VP3, NSP2, and NSP3 along with NSP1.


Subject(s)
Capsid Proteins/metabolism , Host Specificity , Intestines/virology , Rotavirus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Mice , Mice, Inbred BALB C
11.
J Infect Dis ; 207(4): 574-82, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23300162

ABSTRACT

BACKGROUND: Given the high infant measles mortality rate, there is interest in whether a measles immunization regimen beginning at <12 months of age provides lasting immunity. METHODS: Measles-specific immune responses were evaluated in 70 children aged 5-10 years after primary measles vaccine administered at 6, 9, or 12 months. RESULTS: At 5-10 years of age, the stimulation index for measles T-cell proliferation was 11.4 (SE, 1.3), 10.9 (SE, 1.5), and 14.4 (SE 2.1) when the first measles dose was given at 6, 9, or 12 months, respectively. Neutralizing antibody concentration (geometric mean titer [GMT]) in those immunized at 6 months of age was 125 mIU/mL (95% confidence interval [CI], 42-377) in the presence of passive antibodies (PAs) and 335 mIU/mL (95% CI, 211-531) in those without PAs; in those immunized at 9 months, GMTs were 186 mIU/mL (95% CI, 103-335) and 1080 mIU/mL (95% CI, 642-1827) in the presence and absence of PAs, respectively. The GMT was 707 mIU/mL (95% CI, 456-1095) when vaccine was administered at 12 months (P ≤ .04). CONCLUSIONS: Measles-specific T-cell responses were sustained at 5-10 years of age regardless of age at time of primary measles immunization. Neutralizing antibody concentrations were lower in cohorts given the first vaccine dose at 6 months of age and in the presence of PAs; however, responses could be boosted by subsequent doses. Starting measles vaccination at <12 months of age may be beneficial during measles outbreaks or in endemic areas.


Subject(s)
Antibodies, Viral/blood , Measles Vaccine/administration & dosage , Measles Vaccine/immunology , Measles virus/immunology , Measles/immunology , T-Lymphocytes/immunology , Antibodies, Neutralizing/blood , Child , Child, Preschool , Female , Humans , Immunity, Cellular , Immunization Schedule , Lymphocyte Activation , Male , Measles/prevention & control , Vaccination
12.
J Infect Dis ; 201(2): 214-22, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20017631

ABSTRACT

BACKGROUND: The antigen-specific T cell responses of preterm infants to immunization are not well understood. The aim of the present study was to compare the T cell responses of preterm infants after inactivated poliovirus vaccination with those of term infants. METHODS: We prospectively enrolled 2-month-old preterm (gestational age, 33 weeks) and term (gestational age, 37 weeks) infants to receive 3 doses of diphtheria-tetanus toxoids-acellular pertussis-hepatitis B virus-inactivated poliovirus vaccine. Whole blood and peripheral blood mononuclear cells (PBMCs) were stimulated with poliovirus vaccine, and memory T cell activation was analyzed by flow cytometry and lymphoproliferation, respectively. Levels of poliovirus neutralizing antibodies were measured in serum. RESULTS: We enrolled 33 preterm and 50 term infants. Preterm infants had fewer circulating CD4(+)CD45RO(+) memory (P = .005) and CD4(+)CD69(+)IFN-gamma(+) cells activated by staphylococcus enterotoxin B at 2 (P = .015) and 7 (P = .05) months of age. After immunization, preterm and term infants had comparable frequencies of poliovirus-specific CD4(+)CD45RO(+)CD69(+)IFN-gamma(+) memory T cells (P = .79). PBMCs from preterm infants had diminished poliovirus-specific lymphoproliferation (P<.001). Although all infants developed seroprotective poliovirus antibody titers, serotype 1 titers were lower among preterm infants (P = .03). CONCLUSIONS: Preterm infants develop poliovirus-specific T cell responses that are comparable to those of term infants. However, they demonstrate nonspecific and poliovirus-specific functional T cell limitations, suggesting that investigations into whether T cell differences remain as preterm infants mature are warranted.


Subject(s)
Infant, Premature/immunology , Poliovirus Vaccine, Inactivated/immunology , T-Lymphocytes/immunology , Antibodies, Viral/blood , Case-Control Studies , Female , Flow Cytometry , Humans , Immunity, Cellular , Infant , Infant, Newborn , Male , Monocytes/immunology , Prospective Studies , Vaccines, Conjugate/immunology
13.
J Infect Dis ; 200(12): 1921-7, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19909079

ABSTRACT

BACKGROUND: The susceptibility of infants to infections is well defined clinically, and immunologic abnormalities have been described. Immune maturation is complex, however, and the interval during which changes occur during childhood has not been identified. METHODS: To assess age-related differences in the CD4(+) T cell responses, we evaluated the frequency of CD4(+) T cells that produced interferon (IFN) gamma in response to staphylococcal enterotoxin B (SEB) stimulation in 382 healthy infants and children (2 months to 11 years of age) and 66 adults. Flow cytometry was used to assess SEB-induced CD69 and CD40 ligand (CD40-L) expression and IFN-gamma production by CD4(+) and CD45RO(+)CD4(+) T cells. RESULTS: CD69 and CD40-L expression by CD4(+) and CD45RO(+)CD4(+) T cells were similar to adult levels from infancy, but the frequency of activated T cells that produced IFN-gamma remained lower than adult responses until children were 10 years of age. CONCLUSIONS: These observations indicate that the IFN-gamma response of CD4(+) T cells to SEB remains limited for a much longer interval than was reported elsewhere, extending to the second decade of life. Observed differences in CD45RO(+)CD4(+) T cell function indicate that CD4(+) T cells with the same phenotypes do not possess equivalent functional capabilities.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Enterotoxins/immunology , Interferon-gamma/biosynthesis , Adolescent , Adult , Age Factors , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , CD4-Positive T-Lymphocytes/chemistry , CD40 Ligand/analysis , Child , Child, Preschool , Female , Flow Cytometry/methods , Humans , Infant , Lectins, C-Type , Leukocyte Common Antigens/analysis , Lymphocyte Subsets/immunology , Male , Young Adult
14.
Viral Immunol ; 21(2): 163-72, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18419254

ABSTRACT

Understanding the infant host response to measles vaccination is important because of their increased mortality from measles and the need to provide effective protection during the first year of life. Measles-specific T and B-cell responses are lower in infants after measles vaccination than in adults. To define potential mechanisms, we investigated age-related differences in measles-specific T-cell proliferation, CD40-L expression, and IFN-gamma production after measles immunization, and the effects of rhIL-12 and rhIL-15 on these responses. Measles-specific T-cell proliferation and mean IFN-gamma release from infant PBMCs were significantly lower when compared with responses of vaccinated children and adults. Infant responses increased to ranges observed in children and adults when both rhIL-12 and rhIL-15 were added to PBMC cultures. Furthermore, a significant rise in T-cell proliferation and IFN-gamma release was observed when infant PBMCs were stimulated with measles antigen in the presence of rhIL-12 and rhIL-15 compared to measles antigen alone. CD40-L expression by infant and adult T cells stimulated with measles antigen was comparable, but fewer infant CD40-L(+) T cells expressed IFN-gamma. These observations suggest that lower measles-specific T-cell immune responses elicited by measles vaccine in infants may be due to diminished levels of key cytokines.


Subject(s)
Interleukin-12/immunology , Interleukin-15/immunology , Measles Vaccine/immunology , Measles/immunology , Measles/prevention & control , T-Lymphocytes/immunology , Adult , Age Factors , CD40 Ligand/biosynthesis , Cell Proliferation , Cells, Cultured , Child , Child, Preschool , Humans , Infant , Interferon-gamma/biosynthesis , Leukocytes, Mononuclear/immunology , Middle Aged
15.
J Infect Dis ; 197(12): 1669-75, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18419345

ABSTRACT

BACKGROUND: In a mumps outbreak in the United States, many infected individuals were adults who had received 2 doses of mumps vaccine. The persistence of cellular immunity to mumps vaccine has not been defined. METHODS: This was an observational, nonrandomized cohort study evaluating cell-mediated and humoral immunity to mumps in 10 vaccinated and 10 naturally immune adults. Mumps-specific T cell activation and interferon (IFN)-gamma production were measured using lymphoproliferative and flow cytometry assays, and mumps immunoglobulin (Ig) G was measured using enzyme-linked immunosorbent assay. RESULTS: T cell immunity to mumps was high in both groups; 70% of vaccinated and 80% of naturally immune individuals had a positive (> or =3) stimulation index (SI) (P = 1.0). The mean percentages of mumps-specific CD4+ T cells that expressed CD69 and produced IFN-gamma were equivalent in the 2 groups: 0.06% and 0.12%, respectively (P = .11). The mean SIs in the groups were also equivalent, although IFN-gamma concentrations from cultures stimulated with mumps antigen were higher in naturally immune adults than in vaccinated adults (P < or = .01). All adults were positive for mumps IgG. CONCLUSION: T and B cell immunity to mumps was detected in adults at least 10 years after immunization. Except for IFN-gamma release, responses in vaccinated adults paralleled those observed in naturally immune individuals.


Subject(s)
Antibodies, Viral/blood , Mumps Vaccine/immunology , Mumps virus/immunology , Adult , Antibody Formation , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cohort Studies , Flow Cytometry , Humans , Immunity, Cellular , Immunologic Memory , Interferon-gamma/metabolism , Leukocytes, Mononuclear/metabolism , T-Lymphocytes/physiology
16.
Acta Obstet Gynecol Scand ; 86(5): 523-9, 2007.
Article in English | MEDLINE | ID: mdl-17464578

ABSTRACT

BACKGROUND: Neonatal herpes simplex virus (HSV) is a rare but devastating disease. We have conducted pooled analyses of data from 3 cohorts to evaluate the effects of maternal HSV serostatus and HSV type on risk of neonatal HSV acquisition and severity. METHODS: Data from cohorts in Seattle, WA, and Stanford, CA, USA, and Stockholm, Sweden were pooled using Mantel-Haenszel methods. RESULTS: Seventy-eight infants with documented neonatal HSV and known maternal HSV serostatus were included. The risk of neonatal HSV-2 infection was similar in infants born to HSV seronegative women compared with HSV-1 seropositive women (pooled OR: 1.6; 95% CI: 0.6-4.0). The odds of neonatal HSV infection was increased in the presence of exposure to maternal HSV-1 versus HSV-2 (adjusted pooled OR: 19.2; 95% CI: 5.8-63.6). An elevated odds of disseminated HSV in infants born to women with newly acquired genital herpes was observed in Stockholm (OR=13.5; 95% CI: 1.4-630), but not in Seattle or Stanford. CONCLUSION: Our results suggest that maternal HSV-1 antibody offers little, if any, protection against neonatal HSV-2 infection. During reactivation, HSV-1 appears more readily transmissible to the neonate than HSV-2, a concerning finding given the rising frequency of genital HSV-1 infection.


Subject(s)
Herpes Simplex/transmission , Herpes Simplex/virology , Herpesvirus 1, Human , Herpesvirus 2, Human , Infectious Disease Transmission, Vertical , Pregnancy Complications, Infectious/virology , Adult , Antibodies, Viral/blood , Case-Control Studies , Cohort Studies , Databases, Factual , Female , Herpes Simplex/blood , Herpes Simplex/congenital , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/isolation & purification , Herpesvirus 1, Human/pathogenicity , Herpesvirus 2, Human/immunology , Herpesvirus 2, Human/isolation & purification , Herpesvirus 2, Human/pathogenicity , Humans , Infant, Newborn , Pregnancy , Pregnancy Complications, Infectious/blood , Severity of Illness Index , Sweden , United States
17.
Am J Obstet Gynecol ; 195(1): 115-20, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16813750

ABSTRACT

OBJECTIVE: The objective of the study was to assess whether herpes simplex virus antibody avidity is associated with risk of transmission of herpes simplex virus to the neonate. STUDY DESIGN: We developed a novel herpes simplex virus type 1 avidity test based on the commercially available Focus HerpeSelect-1 enzyme-linked immunosorbent assay kit using sera from nonpregnant subjects with genital herpes simplex virus-1 infection. We used this test, and the previously developed herpes simplex virus type 2 avidity test, to compare maternal herpes simplex virus-1 and herpes simplex virus-2 antibody avidity in women who transmitted herpes simplex virus to the neonate and women who had herpes simplex virus isolated from genital secretions at delivery but who did not transmit herpes simplex virus to their infants. RESULTS: Among nonpregnant subjects with genital herpes simplex virus-1 infection whose sera were used to develop the herpes simplex virus-1 avidity test, a significant relationship between herpes simplex virus-1 antibody avidity and time since herpes simplex virus-1 acquisition was observed (P < .001, mixed-effects model), with median avidity values increasing over time after primary infection. Among pregnant, herpes simplex virus-1, or herpes simplex virus-2 seropositive women, 4 of 8 women (50%) with avidity 40 or greater transmitted herpes simplex virus to the neonate, compared with only 12 of 97 (12%) of women with avidity greater than 40 (P = .02). CONCLUSIONS: Herpes simplex virus-1 antibody avidity increased over time after genital herpes simplex virus-1 acquisition, as has been previously observed for herpes simplex virus-2. Among women with herpes simplex virus antibody at delivery, low antibody avidity was associated with herpes simplex virus transmission to the neonate and may be a useful marker for recent seroconversion.


Subject(s)
Antibody Affinity , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Herpesvirus 2, Human/immunology , Pregnancy Complications, Infectious/immunology , Adult , Female , Herpes Simplex/transmission , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical , Pregnancy , ROC Curve , Sensitivity and Specificity , Time Factors
18.
Viral Immunol ; 17(2): 298-307, 2004.
Article in English | MEDLINE | ID: mdl-15279707

ABSTRACT

Vaccination of infants against measles remains of global importance, and proposed new vaccine strategies include the use of measles proteins or synthetic peptides as immunogens. We studied cell-mediated immunity to whole measles antigen and measles proteins in immune adults and infants after measles vaccine. Further, we measured CD8+ T cell responses to peptide pools corresponding to the nucelocapsid (N) measles protein in adults given measles vaccine. Cell-mediated immune responses to three of four measles proteins were equivalent to those against whole measles antigen in immune adults. Responses to the fusion (F) protein were lower in infants compared to whole measles antigen (p < or = 0.03). Infant responses to both whole measles antigen and the F protein were lower compared with these responses in adults (p < or = 0.001). CD8+ T cell responses to N peptide pools varied, and differed between immune HLA-A2-positive individuals compared with naive and HLA-A2-negative subjects after measles vaccination. The measles-specific T cell adaptive response of infants is limited compared to adults, including responses to the F protein.


Subject(s)
Antigens, Viral/immunology , Lymphocyte Activation/immunology , Measles Vaccine/immunology , T-Lymphocytes/immunology , Adult , Antibodies, Viral/biosynthesis , Antibodies, Viral/physiology , CD8-Positive T-Lymphocytes/immunology , Humans , Infant , Infant, Newborn , Lymphocyte Activation/drug effects , Measles Vaccine/administration & dosage , Measles Vaccine/pharmacology , Peptides/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/pharmacology
19.
J Infect Dis ; 190(1): 83-90, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15195246

ABSTRACT

BACKGROUND: Shifts in peak measles incidence to children <12 months old and the associated high mortality support the study of an early 2-dose measles vaccine regimen. METHODS: Fifty-five infants were vaccinated with measles vaccine at age 6 (n=32) or 9 (n=23) months, followed by measles-mumps-rubella (MMR)-II vaccine at age 12 months. A control group received MMR-II only at age 12 months. Measles-specific humoral and cell-mediated immunity were evaluated before, 12 weeks after measles immunization, and 24 weeks after MMR-II. RESULTS: Measles-specific T cell proliferation after both doses of vaccine was equivalent, regardless of age or the presence of passive antibodies. Seroconversion rates, geometric mean titers, and the percentage of infants with antibody titers >120 mIU after the first measles vaccine were lower in infants vaccinated at age 6 months, regardless of the presence of passive antibodies, but measles humoral responses increased after the administration of MMR-II vaccine in children initially vaccinated at age 6 or 9 months. CONCLUSION: Measles vaccination elicits T cell responses in infants as young as 6 months old, which may prime the humoral response to the second dose. Initiating measles vaccination as an early 2-dose regimen results in an immunologic response that is likely to have clinical benefits in developed and developing countries.


Subject(s)
Antibodies, Viral/blood , Measles Vaccine/administration & dosage , Measles virus/immunology , Measles-Mumps-Rubella Vaccine/administration & dosage , Measles/prevention & control , T-Lymphocytes/immunology , Humans , Immunization Schedule , Infant , Lymphocyte Activation , Measles/immunology , Measles Vaccine/immunology , Measles-Mumps-Rubella Vaccine/immunology , Treatment Outcome , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...