Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
mBio ; 13(3): e0040122, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35587190

ABSTRACT

Influenza viruses cause respiratory tract infections, which lead to human disease outbreaks and pandemics. Influenza A virus (IAV) circulates in diverse animal species, predominantly aquatic birds. This often results in the emergence of novel viral strains causing severe human disease upon zoonotic transmission. Innate immune sensing of the IAV infection promotes host cell death and inflammatory responses to confer antiviral host defense. Dysregulated respiratory epithelial cell death and excessive proinflammatory responses drive immunopathology in highly pathogenic influenza infections. Here, we discuss the critical mechanisms regulating IAV-induced cell death and proinflammatory responses. We further describe the essential role of the Z-form nucleic acid sensor ZBP1/DAI and RIPK3 in triggering apoptosis, necroptosis, and pyroptosis during IAV infection and their impact on host defense and pathogenicity in vivo. We also discuss the functional importance of ZBP1-RIPK3 signaling in recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other viral infections. Understanding these mechanisms of RNA virus-induced cytopathic and pathogenic inflammatory responses is crucial for targeting pathogenic lung infections and human respiratory illness.


Subject(s)
COVID-19 , Influenza A virus , Influenza, Human , Pneumonia , Animals , Cell Death , Humans , Influenza A virus/physiology , RNA , RNA-Binding Proteins/genetics , SARS-CoV-2
2.
J Leukoc Biol ; 111(2): 497-508, 2022 02.
Article in English | MEDLINE | ID: mdl-34057760

ABSTRACT

Coronaviruses (CoVs) are RNA viruses that cause human respiratory infections. Zoonotic transmission of the SARS-CoV-2 virus caused the recent COVID-19 pandemic, which led to over 2 million deaths worldwide. Elevated inflammatory responses and cytotoxicity in the lungs are associated with COVID-19 severity in SARS-CoV-2-infected individuals. Bats, which host pathogenic CoVs, operate dampened inflammatory responses and show tolerance to these viruses with mild clinical symptoms. Delineating the mechanisms governing these host-specific inflammatory responses is essential to understand host-virus interactions determining the outcome of pathogenic CoV infections. Here, we describe the essential role of inflammasome activation in determining COVID-19 severity in humans and innate immune tolerance in bats that host several pathogenic CoVs. We further discuss mechanisms leading to inflammasome activation in human SARS-CoV-2 infection and how bats are molecularly adapted to suppress these inflammasome responses. We also report an analysis of functionally important residues of inflammasome components that provide new clues of bat strategies to suppress inflammasome signaling and innate immune responses. As spillover of bat viruses may cause the emergence of new human disease outbreaks, the inflammasome regulation in bats and humans likely provides specific strategies to combat the pathogenic CoV infections.


Subject(s)
COVID-19/pathology , Immune Tolerance , Immunity, Innate , Inflammasomes/immunology , SARS-CoV-2/immunology , Animals , COVID-19/immunology , COVID-19/virology , Chiroptera , Humans , Inflammasomes/metabolism , Phylogeny
3.
J Immunol ; 207(10): 2411-2416, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34663620

ABSTRACT

Programmed cell death (PCD) is essential for the innate immune response, which serves as the first line of defense against pathogens. Caspases regulate PCD, immune responses, and homeostasis. Caspase-8 specifically plays multifaceted roles in PCD pathways including pyroptosis, apoptosis, and necroptosis. However, because caspase-8-deficient mice are embryonically lethal, little is known about how caspase-8 coordinates different PCD pathways under physiological conditions. Here, we report an anti-inflammatory role of caspase-8 during influenza A virus infection. We generated viable mice carrying an uncleavable version of caspase-8 (Casp8 DA/DA). We demonstrated that caspase-8 autoprocessing was responsible for activating caspase-3, thereby suppressing gasdermin D-mediated pyroptosis and inflammatory cytokine release. We also found that apoptotic and pyroptotic pathways were activated at the same time during influenza A virus infection, which enabled the cell-intrinsic anti-inflammatory function of the caspase-8-caspase-3 axis. Our findings provide new insight into the immunological consequences of caspase-8-coordinated PCD cross-talk under physiological conditions.


Subject(s)
Caspase 3/immunology , Caspase 8/immunology , Intracellular Signaling Peptides and Proteins/immunology , Orthomyxoviridae Infections/immunology , Phosphate-Binding Proteins/immunology , Animals , Caspase 3/metabolism , Caspase 8/metabolism , Cell Death , Cytokines , Influenza A virus/immunology , Influenza A virus/metabolism , Mice , Orthomyxoviridae Infections/metabolism
4.
J Biol Chem ; 296: 100579, 2021.
Article in English | MEDLINE | ID: mdl-33766561

ABSTRACT

Viruses and hosts have coevolved for millions of years, leading to the development of complex host-pathogen interactions. Influenza A virus (IAV) causes severe pulmonary pathology and is a recurrent threat to human health. Innate immune sensing of IAV triggers a complex chain of host responses. IAV has adapted to evade host defense mechanisms, and the host has coevolved to counteract these evasion strategies. However, the molecular mechanisms governing the balance between host defense and viral immune evasion is poorly understood. Here, we show that the host protein DEAD-box helicase 3 X-linked (DDX3X) is critical to orchestrate a multifaceted antiviral innate response during IAV infection, coordinating the activation of the nucleotide-binding oligomerization domain-like receptor with a pyrin domain 3 (NLRP3) inflammasome, assembly of stress granules, and type I interferon (IFN) responses. DDX3X activated the NLRP3 inflammasome in response to WT IAV, which carries the immune evasive nonstructural protein 1 (NS1). However, in the absence of NS1, DDX3X promoted the formation of stress granules that facilitated efficient activation of type I IFN signaling. Moreover, induction of DDX3X-containing stress granules by external stimuli after IAV infection led to increased type I IFN signaling, suggesting that NS1 actively inhibits stress granule-mediated host responses and DDX3X-mediated NLRP3 activation counteracts this action. Furthermore, the loss of DDX3X expression in myeloid cells caused severe pulmonary pathogenesis and morbidity in IAV-infected mice. Together, our findings show that DDX3X orchestrates alternate modes of innate host defense which are critical to fight against NS1-mediated immune evasion strategies during IAV infection.


Subject(s)
DEAD-box RNA Helicases/metabolism , Immunity, Innate , Inflammasomes/metabolism , Influenza A virus/physiology , Interferon Type I/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Influenza A virus/immunology , Mice
5.
Crit Rev Immunol ; 41(3): 43-56, 2021.
Article in English | MEDLINE | ID: mdl-35378010

ABSTRACT

Pathogenic coronaviruses (CoVs) have caused human respiratory infections and severe disease outbreaks in the past two decades. Recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans shows high transmissibility causing a wide range of clinical outcomes, named coronavirus disease-2019 (COVID-19), which emerged into an ongoing pandemic. Innate immune sensing of SARS-CoV-2 infection is critical for mounting antiviral and inflammatory responses to restrict the viral spread and initiate lung tissue repair processes. However, excessive cytokine and chemokine levels and dysregulated inflammatory immune cell function in the lungs are associated with respiratory failure and severe COVID-19. Thus, there is a tremendous need for understanding SARS-CoV-2-host interactions determining the aberrant inflammatory responses and loss of respiratory function. In this article, we discuss host innate immune responses determining dysregulated inflammation and immunopathology during SARS-CoV-2 infection. We also provide the perspective for the inflammatory cell death contribution for this immunopathology. Virus-induced acute host responses are complex, and elucidating this complex mechanism facilitates safe therapeutic interventions to alleviate inflammation-mediated immunopathology during pathogenic virus infections.


Subject(s)
COVID-19 , Immunity, Innate , Inflammation , SARS-CoV-2 , Cell Death , Cytokines/metabolism , Humans , Lung , SARS-CoV-2/pathogenicity
6.
Immunohorizons ; 4(12): 789-796, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33310881

ABSTRACT

Bacterial pathogens from the genus Yersinia cause fatal sepsis and gastritis in humans. Innate immune signaling and inflammatory cell death (pyroptosis, apoptosis, and necroptosis [PANoptosis]) serve as a first line of antimicrobial host defense. The receptor-interacting protein kinase 1 (RIPK1) is essential for Yersinia-induced pyroptosis and apoptosis and an effective host response. However, it is not clear whether RIPK1 assembles a multifaceted cell death complex capable of regulating caspase-dependent pyroptosis and apoptosis or whether there is cross-talk with necroptosis under these conditions. In this study, we report that Yersinia activates PANoptosis, as evidenced by the concerted activation of proteins involved in PANoptosis. Genetic deletion of RIPK1 abrogated the Yersinia-induced activation of the inflammasome/pyroptosis and apoptosis but enhanced necroptosis. We also found that Yersinia induced assembly of a RIPK1 PANoptosome complex capable of regulating all three branches of PANoptosis. Overall, our results demonstrate a role for the RIPK1 PANoptosome in Yersinia-induced inflammatory cell death and host defense.


Subject(s)
Inflammation/pathology , Necroptosis , Pyroptosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Yersinia pseudotuberculosis/pathogenicity , Animals , Inflammasomes , Inflammation/etiology , Inflammation/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction
7.
J Biol Chem ; 295(52): 18276-18283, 2020 12 25.
Article in English | MEDLINE | ID: mdl-33109609

ABSTRACT

Candida albicans and Aspergillus fumigatus are dangerous fungal pathogens with high morbidity and mortality, particularly in immunocompromised patients. Innate immune-mediated programmed cell death (pyroptosis, apoptosis, necroptosis) is an integral part of host defense against pathogens. Inflammasomes, which are canonically formed upstream of pyroptosis, have been characterized as key mediators of fungal sensing and drivers of proinflammatory responses. However, the specific cell death pathways and key upstream sensors activated in the context of Candida and Aspergillus infections are unknown. Here, we report that C. albicans and A. fumigatus infection induced inflammatory programmed cell death in the form of pyroptosis, apoptosis, and necroptosis (PANoptosis). Further, we identified the innate immune sensor Z-DNA binding protein 1 (ZBP1) as the apical sensor of fungal infection responsible for activating the inflammasome/pyroptosis, apoptosis, and necroptosis. The Zα2 domain of ZBP1 was required to promote this inflammasome activation and PANoptosis. Overall, our results demonstrate that C. albicans and A. fumigatus induce PANoptosis and that ZBP1 plays a vital role in inflammasome activation and PANoptosis in response to fungal pathogens.


Subject(s)
Apoptosis , Fungi/pathogenicity , Inflammation/pathology , Necroptosis , Pyroptosis , RNA-Binding Proteins/metabolism , Animals , Humans , Inflammasomes , Inflammation/etiology , Inflammation/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , RNA-Binding Proteins/genetics
8.
Article in English | MEDLINE | ID: mdl-32547960

ABSTRACT

Programmed cell death plays crucial roles in organismal development and host defense. Recent studies have highlighted mechanistic overlaps and extensive, multifaceted crosstalk between pyroptosis, apoptosis, and necroptosis, three programmed cell death pathways traditionally considered autonomous. The growing body of evidence, in conjunction with the identification of molecules controlling the concomitant activation of all three pathways by pathological triggers, has led to the development of the concept of PANoptosis. During PANoptosis, inflammatory cell death occurs through the collective activation of pyroptosis, apoptosis, and necroptosis, which can circumvent pathogen-mediated inhibition of individual death pathways. Many of the molecular details of this emerging pathway are unclear. Here, we describe the activation of PANoptosis by bacterial and viral triggers and report protein interactions that reveal the formation of a PANoptosome complex. Infection of macrophages with influenza A virus, vesicular stomatitis virus, Listeria monocytogenes, or Salmonella enterica serovar Typhimurium resulted in robust cell death and the hallmarks of PANoptosis activation. Combined deletion of the PANoptotic components caspase-1 (CASP1), CASP11, receptor-interacting serine/threonine-protein kinase 3 (RIPK3), and CASP8 largely protected macrophages from cell death induced by these pathogens, while deletion of individual components provided reduced or no protection. Further, molecules from the pyroptotic, apoptotic, and necroptotic cell death pathways interacted to form a single molecular complex that we have termed the PANoptosome. Overall, our study identifies pathogens capable of activating PANoptosis and the formation of a PANoptosome complex.


Subject(s)
Apoptosis , Necroptosis , Pyroptosis , Animals , Caspase 1 , Caspase 8 , Caspases, Initiator , Influenza A virus , Listeria monocytogenes , Macrophages , Mice , Receptor-Interacting Protein Serine-Threonine Kinases , Salmonella typhimurium , Vesicular stomatitis Indiana virus
9.
J Exp Med ; 217(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32584411

ABSTRACT

ZBP1 triggers NLRP3 inflammasome activation/pyroptosis, apoptosis, and necroptosis; the specific ligand for ZBP1 activation remains ambiguous. Recent studies, including Devos et al. in this issue of JEM (https://doi.org/10.1084/jem.20191913), collectively suggest that ZBP1 sensing Z-nucleic acids is critical for cell death/inflammatory disease.


Subject(s)
Necroptosis , Nucleic Acids , Biology , Humans , Inflammation , Keratinocytes , RNA-Binding Proteins
10.
J Biol Chem ; 295(24): 8325-8330, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32350114

ABSTRACT

Z-DNA-binding protein 1 (ZBP1) is an innate immune sensor of nucleic acids that regulates host defense responses and development. ZBP1 activation triggers inflammation and pyroptosis, necroptosis, and apoptosis (PANoptosis) by activating receptor-interacting Ser/Thr kinase 3 (RIPK3), caspase-8, and the NLRP3 inflammasome. ZBP1 is unique among innate immune sensors because of its N-terminal Zα1 and Zα2 domains, which bind to nucleic acids in the Z-conformation. However, the specific role of these Zα domains in orchestrating ZBP1 activation and subsequent inflammation and cell death is not clear. Here we generated Zbp1ΔZα2/ΔZα2 mice that express ZBP1 lacking the Zα2 domain and demonstrate that this domain is critical for influenza A virus-induced PANoptosis and underlies perinatal lethality in mice in which the RIP homotypic interaction motif domain of RIPK1 has been mutated (Ripk1mRHIM/mRHIM). Deletion of the Zα2 domain in ZBP1 abolished influenza A virus-induced PANoptosis and NLRP3 inflammasome activation. Furthermore, deletion of the Zα2 domain of ZBP1 was sufficient to rescue Ripk1mRHIM/mRHIM mice from perinatal lethality caused by ZBP1-driven cell death and inflammation. Our findings identify the essential role of the Zα2 domain of ZBP1 in several physiological functions and establish a link between Z-RNA sensing via the Zα2 domain and promotion of influenza-induced PANoptosis and perinatal lethality.


Subject(s)
Embryo Loss/pathology , Embryonic Development , Necroptosis , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Pyroptosis , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Humans , Inflammasomes/metabolism , Influenza, Human/metabolism , Influenza, Human/pathology , Mice, Inbred C57BL , Protein Domains , Sequence Deletion
11.
Annu Rev Immunol ; 38: 567-595, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32017655

ABSTRACT

Caspases are a family of conserved cysteine proteases that play key roles in programmed cell death and inflammation. In multicellular organisms, caspases are activated via macromolecular signaling complexes that bring inactive procaspases together and promote their proximity-induced autoactivation and proteolytic processing. Activation of caspases ultimately results in programmed execution of cell death, and the nature of this cell death is determined by the specific caspases involved. Pioneering new research has unraveled distinct roles and cross talk of caspases in the regulation of programmed cell death, inflammation, and innate immune responses. In-depth understanding of these mechanisms is essential to foster the development of precise therapeutic targets to treat autoinflammatory disorders, infectious diseases, and cancer. This review focuses on mechanisms governing caspase activation and programmed cell death with special emphasis on the recent progress in caspase cross talk and caspase-driven gasdermin D-induced pyroptosis.


Subject(s)
Caspases/metabolism , Cell Death , Inflammation/etiology , Inflammation/metabolism , Neoplasm Proteins/genetics , Pyroptosis/genetics , Animals , Apoptosis , Biomarkers , Caspases/genetics , Cell Death/genetics , Disease Susceptibility , Enzyme Activation , Humans , Inflammation/pathology , Neoplasm Proteins/metabolism , Signal Transduction
12.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31869420

ABSTRACT

RIPK1 kinase activity has been shown to be essential to driving pyroptosis, apoptosis, and necroptosis. However, here we show a kinase activity-independent role for RIPK1 in these processes using a model of TLR priming in a TAK1-deficient setting to mimic pathogen-induced priming and inhibition. TLR priming of TAK1-deficient macrophages triggered inflammasome activation, including the activation of caspase-8 and gasdermin D, and the recruitment of NLRP3 and ASC into a novel RIPK1 kinase activity-independent cell death complex to drive pyroptosis and apoptosis. Furthermore, we found fully functional RIPK1 kinase activity-independent necroptosis driven by the RIPK3-MLKL pathway in TAK1-deficient macrophages. In vivo, TAK1 inactivation resulted in RIPK3-caspase-8 signaling axis-driven myeloid proliferation and a severe sepsis-like syndrome. Overall, our study highlights a previously unknown mechanism for RIPK1 kinase activity-independent inflammasome activation and pyroptosis, apoptosis, and necroptosis (PANoptosis) that could be targeted for treatment of TAK1-associated myeloid proliferation and sepsis.


Subject(s)
Apoptosis/immunology , Immunity, Innate/immunology , Inflammation/immunology , MAP Kinase Kinase Kinases/immunology , Necroptosis/immunology , Pyroptosis/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Caspase 8/immunology , Female , Inflammasomes/immunology , Macrophages/immunology , Mice , Mice, Knockout , Signal Transduction/immunology
13.
Article in English | MEDLINE | ID: mdl-31850239

ABSTRACT

Cell death is central to development, organismal homeostasis, and immune responses. The cell death field has experienced tremendous progress by delineating the molecular programs specific to each of the apoptotic and inflammatory cell death pathways. Moreover, the discovery of the inflammasomes and pyroptosis and necroptosis pathway regulators have provided the genetic basis for the programmed inflammatory cell death pathways. Earlier research highlighted the unique regulation of each of these pathways, but emerging studies discovered co-regulation and crosstalk between these seemingly different cell death complexes. The progress in this area has led to an idea that master regulators play central roles in orchestrating multiple cell death pathways. Here, we provide a brief review of the master regulators, the innate immune sensor ZBP1 and the essential cell survival kinase TAK1, that play vital roles in the regulation of RIPK1/RIPK3-FADD-caspase-8 cell death complex assembly and its versatility in executing Pyroptosis, Apoptosis, and Necroptosis, which we dubbed here as PAN-optosis. Furthermore, we discuss the implications and therapeutic potential of targeting these master regulators in health and disease. One Sentence Summary: ZBP1 and TAK1 regulate PAN-optosis.


Subject(s)
Inflammasomes/metabolism , MAP Kinase Kinase Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA-Binding Proteins/metabolism , Animals , Apoptosis , Disease Susceptibility , Humans , Necroptosis , Pyroptosis
14.
Trends Immunol ; 40(12): 1073-1075, 2019 12.
Article in English | MEDLINE | ID: mdl-31735511

ABSTRACT

A recent study by Rodriguez-Ruiz et al. suggests that inhibition of apoptotic caspases can augment radiation-induced antitumor immunity, independent of type I IFN. Their findings also highlight caspase-independent cytotoxicity in radiation therapy-induced antitumor immunity, proposing SLC7A2 as a new putative prognostic marker for breast cancer.

15.
Nature ; 573(7775): 590-594, 2019 09.
Article in English | MEDLINE | ID: mdl-31511697

ABSTRACT

The cellular stress response has a vital role in regulating homeostasis by modulating cell survival and death. Stress granules are cytoplasmic compartments that enable cells to survive various stressors. Defects in the assembly and disassembly of stress granules are linked to neurodegenerative diseases, aberrant antiviral responses and cancer1-5. Inflammasomes are multi-protein heteromeric complexes that sense molecular patterns that are associated with damage or intracellular pathogens, and assemble into cytosolic compartments known as ASC specks to facilitate the activation of caspase-1. Activation of inflammasomes induces the secretion of interleukin (IL)-1ß and IL-18 and drives cell fate towards pyroptosis-a form of programmed inflammatory cell death that has major roles in health and disease6-12. Although both stress granules and inflammasomes can be triggered by the sensing of cellular stress, they drive contrasting cell-fate decisions. The crosstalk between stress granules and inflammasomes and how this informs cell fate has not been well-studied. Here we show that the induction of stress granules specifically inhibits NLRP3 inflammasome activation, ASC speck formation and pyroptosis. The stress granule protein DDX3X interacts with NLRP3 to drive inflammasome activation. Assembly of stress granules leads to the sequestration of DDX3X, and thereby the inhibition of NLRP3 inflammasome activation. Stress granules and the NLRP3 inflammasome compete for DDX3X molecules to coordinate the activation of innate responses and subsequent cell-fate decisions under stress conditions. Induction of stress granules or loss of DDX3X in the myeloid compartment leads to a decrease in the production of inflammasome-dependent cytokines in vivo. Our findings suggest that macrophages use the availability of DDX3X to interpret stress signals and choose between pro-survival stress granules and pyroptotic ASC specks. Together, our data demonstrate the role of DDX3X in driving NLRP3 inflammasome and stress granule assembly, and suggest a rheostat-like mechanistic paradigm for regulating live-or-die cell-fate decisions under stress conditions.


Subject(s)
Cell Death/genetics , DEAD-box RNA Helicases/metabolism , Inflammasomes/genetics , Macrophages/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Stress, Physiological/genetics , Animals , Cell Line , Cell Survival/genetics , DEAD-box RNA Helicases/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , HEK293 Cells , Humans , Inflammasomes/immunology , Macrophages/immunology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
17.
J Virol ; 92(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29237847

ABSTRACT

Trimeric HIV-1 envelope (Env) immunogens are attractive due to their ability to display quaternary epitopes targeted by broadly neutralizing antibodies (bNAbs) while obscuring unfavorable epitopes. Results from the RV144 trial highlighted the importance of vaccine-induced HIV-1 Env V1V2-directed antibodies, with key regions of the V2 loop as targets for vaccine-mediated protection. We recently reported that a trimeric JRFL-gp120 immunogen, generated by inserting an N-terminal trimerization domain in the V1 loop region of a cyclically permuted gp120 (cycP-gp120), induces neutralizing activity against multiple tier-2 HIV-1 isolates in guinea pigs in a DNA prime/protein boost approach. Here, we tested the immunogenicity of cycP-gp120 in a protein prime/boost approach in rabbits and as a booster immunization to DNA/modified vaccinia Ankara (MVA)-vaccinated rabbits and rhesus macaques. In rabbits, two cycP-gp120 protein immunizations induced 100-fold higher titers of high-avidity gp120-specific IgG than two gp120 immunizations, with four total gp120 immunizations being required to induce comparable titers. cycP-gp120 also induced markedly enhanced neutralizing activity against tier-1A and -1B HIV-1 isolates, substantially higher binding and breadth to gp70-V1V2 scaffolds derived from a multiclade panel of global HIV-1 isolates, and antibodies targeting key regions of the V2-loop region associated with reduced risk of infection in RV144. Similarly, boosting MVA- or DNA/MVA-primed rabbits or rhesus macaques with cycP-gp120 showed a robust expansion of gp70-V1V2-specific IgG, neutralization breadth to tier-1B HIV-1 isolates, and antibody-dependent cellular cytotoxicity activity. These results demonstrate that cycP-gp120 serves as a robust HIV Env immunogen that induces broad anti-V1V2 antibodies and promotes neutralization breadth against HIV-1.IMPORTANCE Recent focus in HIV-1 vaccine development has been the design of trimeric HIV-1 Env immunogens that closely resemble native HIV-1 Env, with a major goal being the induction of bNAbs. While the generation of bNAbs is considered a gold standard in vaccine-induced antibody responses, results from the RV144 trial showed that nonneutralizing antibodies directed toward the V1V2 loop of HIV-1 gp120, specifically the V2 loop region, were associated with decreased risk of infection, demonstrating the need for the development of Env immunogens that induce a broad anti-V1V2 antibody response. In this study, we show that a novel trimeric gp120 protein, cycP-gp120, generates high titers of high-avidity and broadly cross-reactive anti-V1V2 antibodies, a result not found in animals immunized with monomeric gp120. These results reveal the potential of cycP-gp120 as a vaccine candidate to induce antibodies associated with reduced risk of HIV-1 infection in humans.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/blood , HIV Envelope Protein gp120/immunology , HIV Infections/prevention & control , HIV-1/immunology , Immunization/methods , AIDS Vaccines/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibody-Dependent Cell Cytotoxicity , Cross Reactions/immunology , Drug Design , Epitopes/chemistry , Epitopes/immunology , Guinea Pigs , HIV Antibodies/immunology , HIV Antigens/immunology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , Humans , Immunization, Secondary , Immunogenicity, Vaccine , Immunoglobulin G/blood , Macaca mulatta , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/immunology
18.
J Exp Med ; 214(8): 2217-2229, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28634194

ABSTRACT

Innate sensing of influenza virus infection induces activation of programmed cell death pathways. We have recently identified Z-DNA-binding protein 1 (ZBP1) as an innate sensor of influenza A virus (IAV). ZBP1-mediated IAV sensing is critical for triggering programmed cell death in the infected lungs. Surprisingly, little is known about the mechanisms regulating ZBP1 activation to induce programmed cell death. Here, we report that the sensing of IAV RNA by retinoic acid inducible gene I (RIG-I) initiates ZBP1-mediated cell death via the RIG-I-MAVS-IFN-ß signaling axis. IAV infection induces ubiquitination of ZBP1, suggesting potential regulation of ZBP1 function through posttranslational modifications. We further demonstrate that ZBP1 senses viral ribonucleoprotein (vRNP) complexes of IAV to trigger cell death. These findings collectively indicate that ZBP1 activation requires RIG-I signaling, ubiquitination, and vRNP sensing to trigger activation of programmed cell death pathways during IAV infection. The mechanism of ZBP1 activation described here may have broader implications in the context of virus-induced cell death.


Subject(s)
Cell Death/physiology , Glycoproteins/physiology , Ribonucleoproteins/metabolism , Animals , Glycoproteins/metabolism , Influenza A virus/physiology , Membrane Proteins/physiology , Mice, Inbred BALB C , Mice, Knockout , Nerve Tissue Proteins/physiology , Orthomyxoviridae Infections/metabolism , RNA-Binding Proteins , Receptors, Cell Surface , Signal Transduction/physiology , Ubiquitination
19.
Int Immunol ; 29(5): 201-210, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28531279

ABSTRACT

Inflammasomes are multimeric protein complexes that regulate inflammatory responses and pyroptotic cell death to exert host defense against microbes. Intracellular pattern-recognition receptors such as nucleotide-binding domain and leucine-rich repeat receptors (NLRs) and absent in melanoma 2 like receptors (ALRs) assemble the inflammasome complexes in response to pathogens and danger or altered-self signals in the cell. Inflammasome sensors, in association with an adaptor protein-apoptosis-associated speck-like protein containing a caspase-activation and -recruitment domain (ASC)-activate inflammatory caspase-1 to enable the release of inflammatory cytokines and induce cell death, conferring host defense against pathogens. Beyond infectious diseases, the importance of inflammasomes is implicated in a variety of clinical conditions such as auto-inflammatory diseases, neuro-degeneration and metabolic disorders and the development of cancers. Understanding inflammasome activation and its molecular regulation can unveil therapeutic targets for controlling inflammasome-mediated disorders. In this review, we describe recent advances in inflammasome biology and discuss its activation, structural insights into inflammasome assembly and mechanisms for the execution of pyroptosis.


Subject(s)
Inflammasomes/metabolism , Multiprotein Complexes/metabolism , Pyroptosis , Animals , DNA-Binding Proteins/metabolism , Humans , Immunity, Innate , NLR Proteins , Protein Multimerization , Signal Transduction
20.
J Biol Chem ; 292(24): 10197-10219, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28446609

ABSTRACT

The gp120 subunit of the HIV-1 envelope (Env) protein is heavily glycosylated at ∼25 glycosylation sites, of which ∼7-8 are located in the V1/V2 and V3 variable loops and the others in the remaining core gp120 region. Glycans partially shield Env from recognition by the host immune system and also are believed to be indispensable for proper folding of gp120 and for viral infectivity. Previous attempts to alter glycosylation sites in Env typically involved mutating the glycosylated asparagine residues to structurally similar glutamines or alanines. Here, we confirmed that such mutations at multiple glycosylation sites greatly diminish viral infectivity and result in significantly reduced binding to both neutralizing and non-neutralizing antibodies. Therefore, using an alternative approach, we combined evolutionary information with structure-guided design and yeast surface display to produce properly cleaved HIV-1 Env variants that lack all 15 core gp120 glycans, yet retain conformational integrity and multiple-cycle viral infectivity and bind to several broadly neutralizing antibodies (bNAbs), including trimer-specific antibodies and a germline-reverted version of the bNAb VRC01. Our observations demonstrate that core gp120 glycans are not essential for folding, and hence their likely primary role is enabling immune evasion. We also show that our glycan removal approach is not strain restricted. Glycan-deficient Env derivatives can be used as priming immunogens because they should engage and activate a more divergent set of germlines than fully glycosylated Env. In conclusion, these results clarify the role of core gp120 glycosylation and illustrate a general method for designing glycan-free folded protein derivatives.


Subject(s)
HIV Envelope Protein gp120/metabolism , HIV-1/metabolism , Immune Evasion , Models, Molecular , Protein Processing, Post-Translational , Amino Acid Substitution , Antibodies, Neutralizing/metabolism , Antibodies, Viral , Antibody Specificity , Asparagine/metabolism , Glycosylation , HIV Envelope Protein gp120/antagonists & inhibitors , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp41/antagonists & inhibitors , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/metabolism , HIV-1/immunology , HIV-1/pathogenicity , Humans , Mutagenesis, Site-Directed , Mutation , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Engineering , Protein Folding , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...