Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Type of study
Publication year range
1.
Aging (Albany NY) ; 11(13): 4367-4381, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31283526

ABSTRACT

Antibody responses to vaccinations or infections decline upon aging. In this study we tested if metabolic changes in B cells may contribute to attenuation of responses to influenza vaccination in aged humans. Our data show that aging affects mitochondrial functions in B cells leading to increases in mitochondrial reactive oxygen species (MROS) and mitochondrial mass (MM) in some aged B cell subsets and decreases in expression levels of Sirtuin 1 (SIRT1), Forkhead box protein (FOX)O1 and carnitine palmitoyltransferase 1 (CPT-1). Seahorse analyses showed minor defects in glycolysis in the aged B cells after activation but a strong reduction in oxidative phosphorylation. The analyses of the transcriptome revealed further pronounced defects in one-carbon metabolism, a pathway that is essential for amino acid and nucleotide metabolism. Overall our data support the notion that the declining ability of aged B cells to increase their metabolism following activation contributes to the weakened antibody responses of the elderly.


Subject(s)
Aging/immunology , Antibody Formation , B-Lymphocytes/metabolism , Adult , Aged , Aged, 80 and over , Energy Metabolism , Female , Humans , Male
2.
EBioMedicine ; 31: 25-35, 2018 May.
Article in English | MEDLINE | ID: mdl-29685793

ABSTRACT

We report on prime-boost vaccine regimens with two simian adenovirus (Ad) vectors (SAdV) or two human serotype Ad vectors (HAdV) expressing Gag and gp160 of simian immunodeficiency virus (SIV)mac239 tested in HAdV-seropositive rhesus macaques (RMs) repeatedly challenged rectally with low doses of SIVmac251. Both vaccine regimens reduced set point and peak viral loads (PVL) and accelerated viral clearance. In SAdV-vaccinated controller genotype RMs resistance against infection correlated with levels of envelope (Env)-specific antibody (Ab) titers. In both vaccine groups CD8+T cells controlled viral loads (VL) upon infection. Circulating CD4+ and CD8+ T cells showed significant changes in their transcriptome over time following vaccination, which differed between the vaccine groups. T cells from SIV-resistant RMs had unique transcriptional profiles indicating that both follicular T helper (TFH) cell responses and highly activated CD8+ T cells may play a role in protection.


Subject(s)
Adenoviridae/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Products, env/immunology , Genetic Vectors/immunology , Immunity, Cellular , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Adenoviridae/genetics , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Gene Products, env/genetics , Genetic Vectors/genetics , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/genetics , Vaccination
3.
Vaccine ; 35(30): 3700-3708, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28583307

ABSTRACT

Antibody responses, B cell subset distribution in blood and the blood transcriptome were analyzed in younger and aged human subjects before and after vaccination with the inactivated influenza vaccine. In the aged, but not the younger, individuals we saw a clear difference in antibody titers including those at baseline depending on the time of vaccination and sample collection. Differences in baseline titers in aged individuals treated in the morning or afternoon in turn affected responsiveness to the vaccine. In both younger and aged individuals, the time of sample collection also affected relative numbers of some of the B cell subsets in blood. A global gene expression analysis with whole blood samples from the aged showed small but statistically significant differences depending on the time of sample collection. Our data do not indicate that timing of vaccination affects immune responsiveness of the aged, but rather shows that in clinical influenza vaccine trials timing of collection of samples can have a major and potentially misleading influence on study outcome. In future vaccine trials, timing of vaccination and sample collection should be recorded carefully to allow for its use as a study covariant.


Subject(s)
Aging , Antibodies, Viral/blood , Blood Specimen Collection , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Vaccination , Adult , Aged , B-Lymphocyte Subsets/immunology , Circadian Rhythm , Clinical Trials as Topic , Female , Gene Expression Profiling , Humans , Influenza, Human/immunology , Influenza, Human/prevention & control , Male , Time Factors , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
4.
Hum Gene Ther Methods ; 27(5): 187-196, 2016 10.
Article in English | MEDLINE | ID: mdl-27604324

ABSTRACT

Adenovirus (Ad) is used extensively for construction of viral vectors, most commonly with deletion in its E1 and/or E3 genomic regions. Previously, our attempts to insert envelope proteins (Env) of HIV-1 into such vectors based on chimpanzee-derived Ad (AdC) viruses were thwarted. Here, we describe that genetic instability of an E1- and E3-deleted AdC vector of serotype C6 expressing Env of HIV-1 can be overcome by reinsertion of E3 sequences with anti-apoptotic activities. This partial E3 deletion presumably delays premature death of HEK-293 packaging cell lines due to Env-induced cell apoptosis. The same partial E3 deletion also allows for the generation of stable glycoprotein 140 (gp140)- and gp160-expressing Ad vectors based on AdC7, a distinct AdC serotype. Env-expressing AdC vectors containing the partial E3 deletion are genetically stable upon serial cell culture passaging, produce yields comparable to those of other AdC vectors, and induce transgene product-specific antibody responses in mice. A partial E3 deletion thereby allows expansion of the repertoire of transgenes that can be expressed by Ad vectors.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Genetic Vectors/genetics , env Gene Products, Human Immunodeficiency Virus/biosynthesis , Animals , Genetic Vectors/therapeutic use , HEK293 Cells , HIV-1/genetics , Humans , Mice , Serogroup , Transgenes/genetics , Virus Replication/genetics , env Gene Products, Human Immunodeficiency Virus/therapeutic use
5.
J Leukoc Biol ; 96(5): 821-31, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25082150

ABSTRACT

In this study, we tested the effect of neutralizing Abs to different serotypes of E1-deleted Ad vectors on the immunogenicity of the homologous Ad vector or a vector derived from a heterologous serotype. Our results showed that, as expected, even low titers of passively transferred neutralizing Abs significantly reduced the homologous vectors' ability to elicit transgene-specific CD8(+) T cell responses. In addition, Abs changed the fate of transgene product-specific CD8(+) T cells by promoting their transition into the central memory cell pool, which resulted in markedly enhanced expansion of transgene product-specific CD8(+) T cells after a boost with a heterologous Ad vector. Non-neutralizing Abs specific to a distinct Ad serotype had no effect on the magnitude of transgene product-specific CD8(+) T cells induced by a heterologous Ad vector, nor did such Abs promote induction of more resting memory CD8(+) T cells. These results show that Abs to an Ad vaccine carrier affect not only the magnitude but also the profile of a vector-induced CD8(+) T cell response.


Subject(s)
Adenoviridae/genetics , Adenoviridae/immunology , Antibodies, Viral/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , T-Lymphocyte Subsets/immunology , Transgenes/immunology , Adenoviridae/classification , Adenoviridae Infections/immunology , Adenoviridae Infections/prevention & control , Animals , Antibody Specificity/immunology , Cross Reactions/immunology , Epitopes/immunology , Female , Humans , Immune Sera/immunology , Immunization, Passive , Immunologic Memory , Immunophenotyping , Mice , Phenotype , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/metabolism , Transduction, Genetic , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
6.
Vaccine ; 32(27): 3386-92, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24780251

ABSTRACT

Vaccination is an important tool for enhancing immune responses against mucosal pathogens. Intramuscularly administered adenovirus (Ad) vectors have been demonstrated to be strong inducers of both systemic and mucosal immune responses. Further enhancement of immune responses following Ad vaccination is highly desirable. All-trans retinoic acid (ATRA), a biologically active vitamin A metabolite, has been explored as an adjuvant for primary immune responses following vaccination. In this study, we investigated the effect of ATRA on a heterologous Ad prime boost regimen. ATRA co-administration during priming increased mucosal and systemic antibody responses as well as mucosal but not systemic CD8(+) T cell responses. However, this effect was no longer apparent after boosting regardless of whether ATRA was administered at the time of priming, at the time of boosting, or at both immunizations. Our findings confirm ATRA as an adjuvant for primary immune responses and suggest that the adjuvant effect does not extend to secondary immune responses.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/immunology , Immunity, Mucosal , Tretinoin/immunology , Adenoviridae/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Female , Immunization, Secondary , Mice, Inbred BALB C , Receptors, CCR/metabolism , Spleen/immunology , Vaccines, Synthetic/immunology , gag Gene Products, Human Immunodeficiency Virus/immunology
7.
J Leukoc Biol ; 93(3): 437-48, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23271702

ABSTRACT

In this study, we compared the immunogenicity and protection from repeated low-dose intrarectal SIVmac251 challenge in two groups of vaccinated RMs. Animals were immunized with live SIVmac239, which had been attenuated by a deletion of the nef sequence, or they were vaccinated twice with an E1-deleted AdHu5, expressing SIVmac239gag. The vaccinated animals and a cohort of unvaccinated control animals were then challenged 10 times in weekly intervals with low doses of SIVmac251 given rectally. Our results confirm previous studies showing that whereas SIVΔnef provides some degree of protection against viral acquisition after repeated low-dose rectal SIVmac251 challenges, vaccination with an AdHu5gag vaccine designed to induce only antiviral T cell responses is ineffective. As immunological analyses of prechallenge, vaccine-induced T and B cell responses failed to reveal correlates of protection that distinguished the more susceptible from the more resistant vaccinated animals, we carried out RNA-Seq studies of paired pre- and postvaccination samples to identify transcriptional patterns that correlated with the differences in response. We show that gene expression signatures associated with the delayed SIV infection seen in some AdHu5gag recipients were largely present in prevaccination samples of those animals. In contrast, the responding SIVΔnef-immunized animals showed a predominance of vaccine-induced changes, thus enabling us to define inherited and vaccine-induced gene expression signatures and their associated pathways that may play a role in preventing SIV acquisition.


Subject(s)
B-Lymphocytes/immunology , Immunity, Cellular/drug effects , SAIDS Vaccines/pharmacology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus , T-Lymphocytes/immunology , Vaccination , Animals , B-Lymphocytes/virology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Macaca mulatta , Male , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/prevention & control , T-Lymphocytes/virology
8.
Mol Ther ; 19(11): 2021-30, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21587208

ABSTRACT

Hepatic adeno-associated virus serotype 2 (AAV2)-mediated gene transfer failed to achieve sustained transgene product expression in human subjects. We formulated the hypothesis that rejection of AAV-transduced hepatocytes is caused by AAV capsid-specific CD8(+) T cells that become reactivated upon gene transfer. Although this hypothesis was compatible with clinical data, which showed a rise in circulating AAV capsid-specific T cells following injection of AAV vectors, it did not explain that AAV vectors achieved long-term transgene expression in rhesus macaques, which are naturally infected with AAV serotypes closely related to those of humans. To address this apparent contradiction, we tested human and rhesus macaque samples for AAV capsid-specific T cells by intracellular cytokine staining combined with staining for T-cell subset and differentiation markers. This highly sensitive method, which could provide a tool to monitor adverse T-cell responses in gene transfer trials, showed that AAV capsid-specific CD8(+) and CD4(+) T cells can be detected in blood of naturally infected humans and rhesus macaques. They are present at higher frequencies in rhesus macaques. Furthermore, T cells from humans and rhesus macaques exhibit striking differences in their differentiation status and in their functions, which may explain the disparate duration of AAV-mediated gene transfer in these two species.


Subject(s)
Capsid/immunology , Dependovirus/immunology , Parvoviridae Infections/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Animals , Humans , Immunophenotyping , Macaca mulatta , Middle Aged , T-Lymphocytes/metabolism , Young Adult
9.
Mol Ther ; 19(2): 417-26, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21081905

ABSTRACT

Despite enormous efforts by the scientific community, an effective HIV vaccine remains elusive. To further address to what degree T cells in absence of antibodies may protect against simian immunodeficiency virus (SIV) disease progression, rhesus macaques were vaccinated intramuscularly with a chimpanzee-derived Ad vector (AdC) serotype 6 and then boosted intramuscularly with a serologically distinct AdC vector of serotype 7 both expressing Gag of SIVmac239. Animals were subsequently boosted intramuscularly with a modified vaccinia Ankara (MVA) virus expressing Gag and Tat of the homologous SIV before mucosal challenge with a high dose of SIVmac239 given rectally. Whereas vaccinated animals showed only a modest reduction of viral loads, their overall survival was improved, in association with a substantial protection from the loss of CD4(+) T cells. In addition, the two vaccinated Mamu-A*01(+) macaques controlled viral loads to levels below detection within weeks after challenge. These data strongly suggest that T cells, while unable to affect SIV acquisition upon high-dose rectal infection, can reduce disease progression. Induction of potent T-cell responses should thus remain a component of our efforts to develop an efficacious vaccine to HIV-1.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Animals , Female , Male
10.
Eur J Immunol ; 40(12): 3426-38, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21108465

ABSTRACT

Most studies on E1-deleted adenovirus (Ad) vectors as vaccine carriers for antigens of HIV-1 have focused on induction of central immune responses, although stimulation of mucosal immunity at the genital tract (GT), the primary port of entry of HIV-1, would also be highly desirable. In this study, different immunization protocols using chimpanzee-derived adenoviral (AdC) vectors expressing Gag of HIV-1 clade B given in heterologous prime-boost regimens were tested for induction of systemic and genital immune responses. Although i.n. immunization stimulated CD8(+) T-cell responses that could be detected in the GT, this route induced only marginal cellular responses in systemic tissues and furthermore numbers of Gag-specific CD8(+) T cells contracted sharply within a few weeks. On the contrary, i.m. immunization induced higher and more sustained frequencies of vaccine-induced cells which could be detected in the GT as well as systemic compartments. Antigen-specific CD8(+) T cells could be detected 1 year after immunization in all compartments analyzed. Genital memory cells secreted IFN-γ, expressed high levels of CD103 and their phenotypes were consistent with a state of activation. Taken together, the results presented here show that i.m. vaccination with chimpanzee-derived (simian) adenovirus vectors is a suitable strategy to induce a long-lived genital CD8(+) T-cell response.


Subject(s)
Adenoviruses, Simian/genetics , CD8-Positive T-Lymphocytes/drug effects , Genitalia/immunology , HIV-1/immunology , gag Gene Products, Human Immunodeficiency Virus/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Drug Administration Routes , Female , Genetic Vectors/administration & dosage , Genitalia/drug effects , Genitalia/metabolism , Genitalia/pathology , Immunization , Immunologic Memory/drug effects , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Pan troglodytes , gag Gene Products, Human Immunodeficiency Virus/administration & dosage , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
11.
J Immunol ; 184(10): 5475-84, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20410485

ABSTRACT

Immune responses diminish with age resulting in an increased susceptibility of the elderly to infectious agents and an inability to mount protective immune responses to vaccines. Immunosenescence affects multiple aspects of the immune system, including CD8(+) T cells, which control viral infections and are assumed to prevent the development of cancers. In this study, we tested if CD8(+) T cell responses in aged mice could be enhanced through a vaccine that concomitantly expresses Ag and a molecule that blocks an immunoinhibitory pathway. Specifically, we tested a vaccine based on a replication-defective chimpanzee-derived adenovirus vector expressing the nucleoprotein (NP) of influenza A virus as a fusion protein with the HSV type 1 glycoprotein D, which through binding to the herpes virus entry mediator, blocks the immunoinhibitory herpes virus entry mediator B and T lymphocyte attenuator/CD160 pathways. Our results show that the vaccine expressing a fusion protein of NP and glycoprotein D induces significantly higher NP-specific CD8(+) T cell responses in young and aged mice compared with the vaccine expressing NP only.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Influenza A virus/immunology , Receptors, Tumor Necrosis Factor, Member 14/antagonists & inhibitors , Signal Transduction/immunology , Up-Regulation/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Line , Epitopes, T-Lymphocyte/genetics , Female , Genetic Vectors/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Mice , Mice, Inbred C57BL , Nucleocapsid Proteins , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Member 14/physiology , Recombinant Fusion Proteins/immunology , Signal Transduction/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Viral Core Proteins/genetics , Viral Core Proteins/immunology
12.
J Immunol ; 182(10): 6587-99, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414814

ABSTRACT

In the phase IIb STEP trial an HIV-1 vaccine based on adenovirus (Ad) vectors of the human serotype 5 (AdHu5) not only failed to induce protection but also increased susceptibility to HIV-1 infection in individuals with preexisting neutralizing Abs against AdHu5. The mechanisms underlying the increased HIV-1 acquisition rates have not yet been elucidated. Furthermore, it remains unclear if the lack of the vaccine's efficacy reflects a failure of the concept of T cell-mediated protection against HIV-1 or a product failure of the vaccine. Here, we compared two vaccine regimens based on sequential use of AdHu5 vectors or two different chimpanzee-derived Ad vectors in rhesus macaques that were AdHu5 seropositive or seronegative at the onset of vaccination. Our results show that heterologous booster immunizations with the chimpanzee-derived Ad vectors induced higher T and B cell responses than did repeated immunizations with the AdHu5 vector, especially in AdHu5-preexposed macaques.


Subject(s)
AIDS Vaccines/immunology , Adenoviridae/immunology , Antibodies, Viral/immunology , Genetic Vectors/immunology , Human Immunodeficiency Virus Proteins/immunology , Adenoviridae/genetics , Animals , Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/genetics , Human Immunodeficiency Virus Proteins/genetics , Immunization, Secondary/methods , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lymphocyte Activation/immunology , Macaca mulatta , Pan troglodytes , Polymerase Chain Reaction , T-Lymphocytes/immunology
13.
Vaccine ; 25(1): 109-16, 2007 Jan 02.
Article in English | MEDLINE | ID: mdl-16914235

ABSTRACT

AdC6gag37, an E1-deleted adenovirus recombinant derived from the chimpanzee adenovirus serotype 6 expressing a codon-optimized truncated form of gag of HIV-1, was tested for induction of transgene-specific CD8+ T cell responses upon intranasal or intravaginal immunization of mice. Administration of AdC6gag37 induced gag-specific CD8+ T cells at systemic and mucosal sites. Frequencies of gag-specific CD8+ T cells elicited in the genital tract by intravaginal or intranasal immunizations were substantially increased by intranasal priming followed by intravaginal boosting with the same vector. Additionally, intravaginal immunization with AdC6gag37 increased the amount of gammadelta T cells that could be detected in genital tract.


Subject(s)
AIDS Vaccines/administration & dosage , Adenoviruses, Simian/genetics , CD8-Positive T-Lymphocytes/immunology , Gene Products, gag/immunology , Genetic Vectors , Genitalia, Female/immunology , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Adenoviruses, Simian/immunology , Administration, Intravaginal , Animals , Female , Gene Products, gag/administration & dosage , Gene Products, gag/genetics , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , Immunity, Mucosal , Mice , Mice, Inbred BALB C , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...