Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 7(78): eadd0665, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36563168

ABSTRACT

TNF mediates a variety of biological processes including cellular proliferation, inflammatory responses, and cell death and is therefore associated with numerous pathologies including autoinflammatory diseases and septic shock. The inflammatory and cell death responses to TNF have been studied extensively downstream of TNF-R1 and are believed to rely on the formation of proinflammatory complex I and prodeath complex II, respectively. We recently identified a similar multimeric complex downstream of TLR4, termed the TRIFosome, that regulates inflammation and cell death in response to LPS or Yersinia pseudotuberculosis. We present evidence of a role for the TRIFosome downstream of TNF-R1, independent of TLR3 or TLR4 engagement. Specifically, TNF-induced cell death and inflammation in murine macrophages were driven by the TLR4 adaptor TRIF and the LPS co-receptor CD14, highlighting an important role for these proteins beyond TLR-mediated immune responses. Via immunoprecipitation and visualization of TRIF-specific puncta, we demonstrated TRIF- and CD14-dependent formation of prodeath and proinflammatory complexes in response to TNF. Extending these findings, in a murine TNF-induced sepsis model, TRIF and CD14 deficiency decreased systemic inflammation, reduced organ pathology, and improved survival. The outcome of TRIF activation was cell specific, because TNF-induced lethality was mediated by neutrophils and macrophages responding to TNF in a TRIF-dependent manner. Our findings suggest that in addition to their crucial role in TNF production, myeloid cells are central to TNF toxicity and position TRIF and CD14 as universal components of receptor-mediated immune responses.


Subject(s)
Neutrophils , Receptors, Tumor Necrosis Factor, Type I , Animals , Mice , Adaptor Proteins, Vesicular Transport/metabolism , Inflammation/metabolism , Lipopolysaccharide Receptors , Lipopolysaccharides , Macrophages , Neutrophils/metabolism , Signal Transduction , Toll-Like Receptor 4 , Tumor Necrosis Factors/metabolism
2.
Proc Natl Acad Sci U S A ; 119(24): e2113872119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35666872

ABSTRACT

ZBP1 is widely recognized as a mediator of cell death for its role in initiating necroptotic, apoptotic, and pyroptotic cell death pathways in response to diverse pathogenic infection. Herein, we characterize an unanticipated role for ZBP1 in promoting inflammatory responses to bacterial lipopolysaccharide (LPS) or double-stranded RNA (dsRNA). In response to both stimuli, ZBP1 promotes the timely delivery of RIPK1 to the Toll-like receptor (TLR)3/4 adaptor TRIF and M1-ubiquitination of RIPK1, which sustains activation of inflammatory signaling cascades downstream of RIPK1. Strikingly, ZBP1-mediated regulation of these pathways is important in vivo, as Zbp1−/− mice exhibited resistance to LPS-induced septic shock, revealed by prolonged survival and delayed onset of hypothermia due to decreased inflammatory responses and subsequent cell death. Further findings revealed that ZBP1 promotes sustained inflammatory responses by mediating the kinetics of proinflammatory "TRIFosome" complex formation, thus having a profound impact downstream of TLR activation. Given the well-characterized role of ZBP1 as a viral sensor, our results exemplify previously unappreciated crosstalk between the pathways that regulate host responses to bacteria and viruses, with ZBP1 acting as a crucial bridge between the two.


Subject(s)
Inflammation , Toll-Like Receptor 3 , Toll-Like Receptor 4 , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Inflammation/metabolism , Lipopolysaccharides/toxicity , Mice , RNA, Double-Stranded , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism
3.
Proc Natl Acad Sci U S A ; 117(52): 32880-32882, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33318172

ABSTRACT

In vivo clonal expansion of HIV-infected T cells is an important mechanism of viral persistence. In some cases, clonal expansion is driven by HIV proviral DNA integrated into one of a handful of genes. To investigate this phenomenon in vitro, we infected primary CD4+ T cells with an HIV construct expressing GFP and, after nearly 2 mo of culture and multiple rounds of activation, analyzed the resulting integration site distribution. In each of three replicates from each of two donors, we detected large clusters of integration sites with multiple breakpoints, implying clonal selection. These clusters all mapped to a narrow region within the STAT3 gene. The presence of hybrid transcripts splicing HIV to STAT3 sequences supports a model of LTR-driven STAT3 overexpression as a driver of preferential growth. Thus, HIV integration patterns linked to selective T cell outgrowth can be reproduced in cell culture. The single report of an HIV provirus in a case of AIDS-associated B-cell lymphoma with an HIV provirus in the same part of STAT3 also has implications for HIV-induced malignancy.


Subject(s)
Cell Proliferation , HIV/physiology , Proviruses/physiology , T-Lymphocytes/virology , Virus Integration , Cells, Cultured , Clonal Evolution , DNA, Viral/genetics , HIV/genetics , Humans , Proviruses/genetics , STAT3 Transcription Factor/genetics , T-Lymphocytes/physiology
4.
Gut Microbes ; 12(1): 1-20, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33103561

ABSTRACT

Despite the existing association of gut dysbiosis and T cell inflammation in heart failure (HF), whether and how gut microbes contribute to T cell immune responses, cardiac fibrosis and dysfunction in HF remains largely unexplored. Our objective was to investigate whether gut dysbiosis is induced by cardiac pressure overload, and its effect in T cell activation, adverse cardiac remodeling, and cardiac dysfunction. We used 16S rRNA sequencing of fecal samples and discovered that cardiac pressure overload-induced by transverse aortic constriction (TAC) results in gut dysbiosis, characterized by a reduction of tryptophan and short-chain fatty acids producing bacteria in WT mice, but not in T cell-deficient mice (Tcra-/- ) mice. These changes did not result in T cell activation in the gut or gut barrier disruption. Strikingly, microbiota depletion in WT mice resulted in decreased heart T cell infiltration, decreased cardiac fibrosis, and protection from systolic dysfunction in response to TAC. Spontaneous reconstitution of the microbiota partially reversed these effects. We observed decreased cardiac expression of the Aryl hydrocarbon receptor (AhR) and enzymes associated with tryptophan metabolism in WT mice, but not in Tcra-/- mice, or in mice depleted of the microbiota. These findings demonstrate that cardiac pressure overload induced gut dysbiosis and T cell immune responses contribute to adverse cardiac remodeling, and identify the potential contribution of tryptophan metabolites and the AhR to protection from adverse cardiac remodeling and systolic dysfunction in HF.


Subject(s)
Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Heart Failure/physiopathology , T-Lymphocytes/immunology , Ventricular Pressure/physiology , Ventricular Remodeling/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Disease Models, Animal , Endomyocardial Fibrosis/physiopathology , Fatty Acids, Volatile/metabolism , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Right Ventricular/physiopathology , Inflammation/immunology , Lymphocyte Activation/immunology , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Receptors, Aryl Hydrocarbon/biosynthesis , Tryptophan/metabolism
5.
Science ; 367(6484): 1379-1384, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32193329

ABSTRACT

Cell death and inflammation are interdependent host responses to infection. During pyroptotic cell death, interleukin-1ß (IL-1ß) release occurs through caspase-1 and caspase-11-mediated gasdermin D pore formation. In vivo, responses to lipopolysaccharide (LPS) result in IL-1ß secretion. In vitro, however, murine macrophages require a second "danger signal" for the inflammasome-driven maturation of IL-1ß. Recent reports have shown caspase-8-mediated pyroptosis in LPS-activated macrophages but have provided conflicting evidence regarding the release of IL-1ß under these conditions. Here, to further characterize the mechanism of LPS-induced secretion in vitro, we reveal an important role for cellular FLICE-like inhibitory protein (cFLIP) in the regulation of the inflammatory response. Specifically, we show that deficiency of the long isoform cFLIPL promotes complex II formation, driving pyroptosis, and the secretion of IL-1ß in response to LPS alone.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Electron Transport Complex II/metabolism , Inflammasomes/metabolism , Macrophage Activation , Macrophages/physiology , Pyroptosis , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 1/metabolism , Caspase 8/metabolism , Gene Knockdown Techniques , Interleukin-1beta/metabolism , Lipopolysaccharides/immunology , Macrophages/immunology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
6.
Nature ; 564(7734): E8, 2018 12.
Article in English | MEDLINE | ID: mdl-30397346

ABSTRACT

In Fig. 4b of this Article, the x-axis labels 'PGT121' and 'GS-9620' were inadvertently swapped in both graphs. In Fig. 5a, b, 'TLR7' should have been 'GS-9620'. These figures have been corrected online.

7.
Nature ; 563(7731): 360-364, 2018 11.
Article in English | MEDLINE | ID: mdl-30283138

ABSTRACT

The latent viral reservoir is the critical barrier for the development of a cure for HIV-1 infection. Previous studies have shown direct antiviral activity of potent HIV-1 Env-specific broadly neutralizing antibodies (bNAbs) administered when antiretroviral therapy (ART) was discontinued, but it remains unclear whether bNAbs can target the viral reservoir during ART. Here we show that administration of the V3 glycan-dependent bNAb PGT121 together with the Toll-like receptor 7 (TLR7) agonist vesatolimod (GS-9620) during ART delayed viral rebound following discontinuation of ART in simian-human immunodeficiency virus (SHIV)-SF162P3-infected rhesus monkeys in which ART was initiated during early acute infection. Moreover, in the subset of monkeys that were treated with both PGT121 and GS-9620 and that did not show viral rebound after discontinuation of ART, adoptive transfer studies and CD8-depletion studies also did not reveal virus. These data demonstrate the potential of bNAb administration together with innate immune stimulation as a possible strategy for targeting the viral reservoir.


Subject(s)
Antibodies, Viral/immunology , HIV-1/drug effects , HIV-1/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology , Toll-Like Receptor 7/agonists , Adoptive Transfer , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Antibodies, Neutralizing/immunology , CD8 Antigens/deficiency , CD8 Antigens/immunology , DNA, Viral/analysis , Female , HIV Antibodies/immunology , HIV-1/genetics , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , Male , Pteridines/pharmacology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Toll-Like Receptor 7/immunology , Viral Load
8.
Lancet ; 392(10143): 232-243, 2018 07 21.
Article in English | MEDLINE | ID: mdl-30047376

ABSTRACT

BACKGROUND: More than 1·8 million new cases of HIV-1 infection were diagnosed worldwide in 2016. No licensed prophylactic HIV-1 vaccine exists. A major limitation to date has been the lack of direct comparability between clinical trials and preclinical studies. We aimed to evaluate mosaic adenovirus serotype 26 (Ad26)-based HIV-1 vaccine candidates in parallel studies in humans and rhesus monkeys to define the optimal vaccine regimen to advance into clinical efficacy trials. METHODS: We conducted a multicentre, randomised, double-blind, placebo-controlled phase 1/2a trial (APPROACH). Participants were recruited from 12 clinics in east Africa, South Africa, Thailand, and the USA. We included healthy, HIV-1-uninfected participants (aged 18-50 years) who were considered at low risk for HIV-1 infection. We randomly assigned participants to one of eight study groups, stratified by region. Participants and investigators were blinded to the treatment allocation throughout the study. We primed participants at weeks 0 and 12 with Ad26.Mos.HIV (5 × 1010 viral particles per 0·5 mL) expressing mosaic HIV-1 envelope (Env)/Gag/Pol antigens and gave boosters at weeks 24 and 48 with Ad26.Mos.HIV or modified vaccinia Ankara (MVA; 108 plaque-forming units per 0·5 mL) vectors with or without high-dose (250 µg) or low-dose (50 µg) aluminium adjuvanted clade C Env gp140 protein. Those in the control group received 0·9% saline. All study interventions were administered intramuscularly. Primary endpoints were safety and tolerability of the vaccine regimens and Env-specific binding antibody responses at week 28. Safety and immunogenicity were also assessed at week 52. All participants who received at least one vaccine dose or placebo were included in the safety analysis; immunogenicity was analysed using the per-protocol population. We also did a parallel study in rhesus monkeys (NHP 13-19) to assess the immunogenicity and protective efficacy of these vaccine regimens against a series of six repetitive, heterologous, intrarectal challenges with a rhesus peripheral blood mononuclear cell-derived challenge stock of simian-human immunodeficiency virus (SHIV-SF162P3). The APPROACH trial is registered with ClinicalTrials.gov, number NCT02315703. FINDINGS: Between Feb 24, 2015, and Oct 16, 2015, we randomly assigned 393 participants to receive at least one dose of study vaccine or placebo in the APPROACH trial. All vaccine regimens demonstrated favourable safety and tolerability. The most commonly reported solicited local adverse event was mild-to-moderate pain at the injection site (varying from 69% to 88% between the different active groups vs 49% in the placebo group). Five (1%) of 393 participants reported at least one grade 3 adverse event considered related to the vaccines: abdominal pain and diarrhoea (in the same participant), increased aspartate aminotransferase, postural dizziness, back pain, and malaise. The mosaic Ad26/Ad26 plus high-dose gp140 boost vaccine was the most immunogenic in humans; it elicited Env-specific binding antibody responses (100%) and antibody-dependent cellular phagocytosis responses (80%) at week 52, and T-cell responses at week 50 (83%). We also randomly assigned 72 rhesus monkeys to receive one of five different vaccine regimens or placebo in the NHP 13-19 study. Ad26/Ad26 plus gp140 boost induced similar magnitude, durability, and phenotype of immune responses in rhesus monkeys as compared with humans and afforded 67% protection against acquisition of SHIV-SF162P3 infection (two-sided Fisher's exact test p=0·007). Env-specific ELISA and enzyme-linked immunospot assay responses were the principal immune correlates of protection against SHIV challenge in monkeys. INTERPRETATION: The mosaic Ad26/Ad26 plus gp140 HIV-1 vaccine induced comparable and robust immune responses in humans and rhesus monkeys, and it provided significant protection against repetitive heterologous SHIV challenges in rhesus monkeys. This vaccine concept is currently being evaluated in a phase 2b clinical efficacy study in sub-Saharan Africa (NCT03060629). FUNDING: Janssen Vaccines & Prevention BV, National Institutes of Health, Ragon Institute of MGH, MIT and Harvard, Henry M Jackson Foundation for the Advancement of Military Medicine, US Department of Defense, and International AIDS Vaccine Initiative.


Subject(s)
AIDS Vaccines/administration & dosage , HIV-1/immunology , AIDS Vaccines/adverse effects , Abdominal Pain/etiology , Adenoviridae , Adolescent , Adult , Animals , Aspartate Aminotransferases/analysis , Back Pain/etiology , Diarrhea/etiology , Dizziness/etiology , Dose-Response Relationship, Drug , Double-Blind Method , Fatigue/etiology , Genetic Vectors , Healthy Volunteers , Humans , Immunity, Cellular , Immunity, Humoral , Macaca mulatta , Middle Aged , Young Adult
9.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29298888

ABSTRACT

Human and chimpanzee adenovirus vectors are being developed to circumvent preexisting antibodies against common adenovirus vectors such as Ad5. However, baseline immunity to these vectors still exists in human populations. Traditional cloning of new adenovirus vaccine vectors is a long and cumbersome process that takes 2 months or more and that requires rare unique restriction enzyme sites. Here we describe a novel, restriction enzyme-independent method for rapid cloning of new adenovirus vaccine vectors that reduces the total cloning procedure to 1 week. We developed 14 novel adenovirus vectors from rhesus monkeys that can be grown to high titers and that are immunogenic in mice. All vectors grouped with the unusual adenovirus species G and show extremely low seroprevalence in humans. Rapid cloning of novel adenovirus vectors is a promising approach for the development of new vector platforms. Rhesus adenovirus vectors may prove useful for clinical development.IMPORTANCE To overcome baseline immunity to human and chimpanzee adenovirus vectors, we developed 14 novel adenovirus vectors from rhesus monkeys. These vectors are immunogenic in mice and show extremely low seroprevalence in humans. Rhesus adenovirus vectors may prove useful for clinical development.


Subject(s)
Adenoviridae , Adenovirus Vaccines , Cloning, Molecular , Genetic Vectors , Immunogenicity, Vaccine/genetics , A549 Cells , Adenoviridae/genetics , Adenoviridae/immunology , Adenovirus Vaccines/genetics , Adenovirus Vaccines/immunology , Animals , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Macaca mulatta , Mice
10.
Sci Transl Med ; 9(420)2017 Dec 13.
Article in English | MEDLINE | ID: mdl-29237759

ABSTRACT

An effective Zika virus (ZIKV) vaccine will require long-term durable protection. Several ZIKV vaccine candidates have demonstrated protective efficacy in nonhuman primates, but these studies have typically involved ZIKV challenge shortly after vaccination at peak immunity. We show that a single immunization with an adenovirus vector-based vaccine, as well as two immunizations with a purified inactivated virus vaccine, afforded robust protection against ZIKV challenge in rhesus monkeys at 1 year after vaccination. In contrast, two immunizations with an optimized DNA vaccine, which provided complete protection at peak immunity, resulted in reduced protective efficacy at 1 year that was associated with declining neutralizing antibody titers to subprotective levels. These data define a microneutralization log titer of 2.0 to 2.1 as the threshold required for durable protection against ZIKV challenge in this model. Moreover, our findings demonstrate that protection against ZIKV challenge in rhesus monkeys is possible for at least 1 year with a single-shot vaccine.


Subject(s)
Viral Vaccines/immunology , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/immunology , Animals , Antigens, Viral/immunology , DNA, Viral/metabolism , Female , Macaca mulatta , Mice, Inbred BALB C , Vaccination
11.
PLoS Pathog ; 13(8): e1006487, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28771605

ABSTRACT

Studies have demonstrated cross-reactivity of anti-dengue virus (DENV) antibodies in human sera against Zika virus (ZIKV), promoting increased ZIKV infection in vitro. However, the correlation between in vitro and in vivo findings is not well characterized. Thus, we evaluated the impact of heterotypic flavivirus immunity on ZIKV titers in biofluids of rhesus macaques. Animals previously infected (≥420 days) with DENV2, DENV4, or yellow fever virus were compared to flavivirus-naïve animals following infection with a Brazilian ZIKV strain. Sera from DENV-immune macaques demonstrated cross-reactivity with ZIKV by antibody-binding and neutralization assays prior to ZIKV infection, and promoted increased ZIKV infection in cell culture assays. Despite these findings, no significant differences between flavivirus-naïve and immune animals were observed in viral titers, neutralizing antibody levels, or immune cell kinetics following ZIKV infection. These results indicate that prior infection with heterologous flaviviruses neither conferred protection nor increased observed ZIKV titers in this non-human primate ZIKV infection model.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Flavivirus Infections/immunology , Zika Virus Infection/immunology , Animals , Cross Reactions/immunology , Enzyme-Linked Immunosorbent Assay , Flavivirus/immunology , Flavivirus Infections/pathology , Macaca mulatta , Polymerase Chain Reaction , Zika Virus/immunology , Zika Virus Infection/pathology
12.
Cell ; 169(4): 610-620.e14, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28457610

ABSTRACT

Zika virus (ZIKV) is associated with severe neuropathology in neonates as well as Guillain-Barré syndrome and other neurologic disorders in adults. Prolonged viral shedding has been reported in semen, suggesting the presence of anatomic viral reservoirs. Here we show that ZIKV can persist in cerebrospinal fluid (CSF) and lymph nodes (LN) of infected rhesus monkeys for weeks after virus has been cleared from peripheral blood, urine, and mucosal secretions. ZIKV-specific neutralizing antibodies correlated with rapid clearance of virus in peripheral blood but remained undetectable in CSF for the duration of the study. Viral persistence in both CSF and LN correlated with upregulation of mechanistic target of rapamycin (mTOR), proinflammatory, and anti-apoptotic signaling pathways, as well as downregulation of extracellular matrix signaling pathways. These data raise the possibility that persistent or occult neurologic and lymphoid disease may occur following clearance of peripheral virus in ZIKV-infected individuals.


Subject(s)
Zika Virus Infection/immunology , Zika Virus Infection/virology , Animals , Cerebrospinal Fluid/virology , Inflammation/immunology , Lower Gastrointestinal Tract/virology , Lymph Nodes/virology , Macaca mulatta , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
13.
Nature ; 540(7632): 284-287, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27841870

ABSTRACT

The development of immunologic interventions that can target the viral reservoir in HIV-1-infected individuals is a major goal of HIV-1 research. However, little evidence exists that the viral reservoir can be sufficiently targeted to improve virologic control following discontinuation of antiretroviral therapy. Here we show that therapeutic vaccination with Ad26/MVA (recombinant adenovirus serotype 26 (Ad26) prime, modified vaccinia Ankara (MVA) boost) and stimulation of TLR7 (Toll-like receptor 7) improves virologic control and delays viral rebound following discontinuation of antiretroviral therapy in SIV-infected rhesus monkeys that began antiretroviral therapy during acute infection. Therapeutic vaccination with Ad26/MVA resulted in a marked increase in the magnitude and breadth of SIV-specific cellular immune responses in virologically suppressed, SIV-infected monkeys. TLR7 agonist administration led to innate immune stimulation and cellular immune activation. The combination of Ad26/MVA vaccination and TLR7 stimulation resulted in decreased levels of viral DNA in lymph nodes and peripheral blood, and improved virologic control and delayed viral rebound following discontinuation of antiretroviral therapy. The breadth of cellular immune responses correlated inversely with set point viral loads and correlated directly with time to viral rebound. These data demonstrate the potential of therapeutic vaccination combined with innate immune stimulation as a strategy aimed at a functional cure for HIV-1 infection.


Subject(s)
Adenoviridae/genetics , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Immunodeficiency Virus/immunology , Toll-Like Receptor 7/immunology , Vaccinia virus/genetics , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Anti-Retroviral Agents/administration & dosage , DNA, Viral/analysis , DNA, Viral/blood , Female , Genetic Vectors/genetics , HIV Infections/immunology , HIV Infections/therapy , Immunity, Cellular , Immunity, Innate , Macaca mulatta , Male , RNA, Viral/analysis , RNA, Viral/blood , SAIDS Vaccines/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/isolation & purification , Time Factors , Toll-Like Receptor 7/genetics , Viral Load/immunology
14.
Science ; 353(6304): 1129-32, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27492477

ABSTRACT

Zika virus (ZIKV) is responsible for a major ongoing epidemic in the Americas and has been causally associated with fetal microcephaly. The development of a safe and effective ZIKV vaccine is therefore an urgent global health priority. Here we demonstrate that three different vaccine platforms protect against ZIKV challenge in rhesus monkeys. A purified inactivated virus vaccine induced ZIKV-specific neutralizing antibodies and completely protected monkeys against ZIKV strains from both Brazil and Puerto Rico. Purified immunoglobulin from vaccinated monkeys also conferred passive protection in adoptive transfer studies. A plasmid DNA vaccine and a single-shot recombinant rhesus adenovirus serotype 52 vector vaccine, both expressing ZIKV premembrane and envelope, also elicited neutralizing antibodies and completely protected monkeys against ZIKV challenge. These data support the rapid clinical development of ZIKV vaccines for humans.


Subject(s)
Immunogenicity, Vaccine , Vaccines, DNA/immunology , Viral Vaccines/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Adenoviridae , Adoptive Transfer , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Brazil , Female , Genetic Vectors , Humans , Immunoglobulins/immunology , Immunoglobulins/isolation & purification , Macaca mulatta , Male , Mice , Mice, Inbred BALB C , Puerto Rico , Vaccines, DNA/administration & dosage , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage
15.
Nature ; 536(7617): 474-8, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27355570

ABSTRACT

Zika virus (ZIKV) is a flavivirus that is responsible for the current epidemic in Brazil and the Americas. ZIKV has been causally associated with fetal microcephaly, intrauterine growth restriction, and other birth defects in both humans and mice. The rapid development of a safe and effective ZIKV vaccine is a global health priority, but very little is currently known about ZIKV immunology and mechanisms of immune protection. Here we show that a single immunization with a plasmid DNA vaccine or a purified inactivated virus vaccine provides complete protection in susceptible mice against challenge with a strain of ZIKV involved in the outbreak in northeast Brazil. This ZIKV strain has recently been shown to cross the placenta and to induce fetal microcephaly and other congenital malformations in mice. We produced DNA vaccines expressing ZIKV pre-membrane and envelope (prM-Env), as well as a series of deletion mutants. The prM-Env DNA vaccine, but not the deletion mutants, afforded complete protection against ZIKV, as measured by absence of detectable viraemia following challenge, and protective efficacy correlated with Env-specific antibody titers. Adoptive transfer of purified IgG from vaccinated mice conferred passive protection, and depletion of CD4 and CD8 T lymphocytes in vaccinated mice did not abrogate this protection. These data demonstrate that protection against ZIKV challenge can be achieved by single-shot subunit and inactivated virus vaccines in mice and that Env-specific antibody titers represent key immunologic correlates of protection. Our findings suggest that the development of a ZIKV vaccine for humans is likely to be achievable.


Subject(s)
Viral Vaccines/immunology , Zika Virus Infection/prevention & control , Zika Virus Infection/virology , Zika Virus/immunology , Adoptive Transfer , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Specificity , Brazil , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Gene Deletion , Humans , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Mice , Microcephaly/complications , Microcephaly/virology , Vaccines, DNA/chemistry , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, Inactivated/chemistry , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/chemistry , Viral Vaccines/genetics , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus Infection/complications , Zika Virus Infection/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...