Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
Methods Mol Biol ; 2635: 185-202, 2023.
Article de Anglais | MEDLINE | ID: mdl-37074664

RÉSUMÉ

Examining inflammasome-associated speck structures is one of the most preferred and easiest ways to evaluate inflammasome activation. Microscopy-based evaluation of specks is preferable, but this approach is time-consuming and limited to small sample sizes. Speck-containing cells can also be quantitated by a flow cytometric method, time of flight inflammasome evaluation (TOFIE). However, TOFIE cannot perform single-cell analysis such as simultaneously visualizing ASC specks and caspase-1 activity, their location, and physical characteristics. Here we describe the application of an imaging flow cytometry-based approach that overcomes these limitations. Inflammasome and Caspase-1 Activity Characterization and Evaluation (ICCE) is a high-throughput, single-cell, rapid image analysis utilizing the Amnis ImageStream X instrument with over 99.5% accuracy. ICCE quantitatively and qualitatively characterizes the frequency, area, and cellular distribution of ASC specks and caspase-1 activity in mouse and human cells.


Sujet(s)
Protéines adaptatrices de signalisation CARD , Inflammasomes , Humains , Animaux , Souris , Inflammasomes/métabolisme , Protéines adaptatrices de signalisation CARD/métabolisme , Macrophages/métabolisme , Microscopie , Caspases , Caspase-1/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine
2.
Front Immunol ; 12: 752482, 2021.
Article de Anglais | MEDLINE | ID: mdl-34745125

RÉSUMÉ

Although considered the ternary inflammasome structure, whether the singular, perinuclear NLRP3:ASC speck is synonymous with the NLRP3 inflammasome is unclear. Herein, we report that the NLRP3:ASC speck is not required for nigericin-induced inflammasome activation but facilitates and maximizes IL-1ß processing. Furthermore, the NLRP3 agonists H2O2 and MSU elicited IL-1ß maturation without inducing specks. Notably, caspase-1 activity is spatially distinct from the speck, occurring at multiple cytoplasmic sites. Additionally, caspase-1 activity negatively regulates speck frequency and speck size, while speck numbers and IL-1ß processing are negatively correlated, cyclical and can be uncoupled by NLRP3 mutations or inhibiting microtubule polymerization. Finally, when specks are present, caspase-1 is likely activated after leaving the speck structure. Thus, the speck is not the NLRP3 inflammasome itself, but is instead a dynamic structure which may amplify the NLRP3 response to weak stimuli by facilitating the formation and release of small NLRP3:ASC complexes which in turn activate caspase-1.


Sujet(s)
Protéines adaptatrices de signalisation CARD , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Cellules cultivées , Humains , Speckles nucléaires
3.
Front Immunol ; 11: 1828, 2020.
Article de Anglais | MEDLINE | ID: mdl-32983094

RÉSUMÉ

The NLRP3 inflammasome is central to host defense and implicated in various inflammatory diseases and conditions. While the favored paradigm of NLRP3 inflammasome activation stipulates a unifying signal intermediate that de-represses NLRP3, this view has not been tested. Further, structures within NLRP3 required for inflammasome activation are poorly defined. Here we demonstrate that while the NLRP3 LRRs are not auto-repressive and are not required for inflammasome activation by all agonists, distinct sequences within the NLRP3 LRRs positively and negatively modulate inflammasome activation by specific ligands. In addition, elements within the HD1/HD2 "hinge" of NLRP3 and the nucleotide-binding domain have contrasting functions depending upon the specific agonists. Further, while NLRP3 1-432 is minimally sufficient for inflammasome activation by all agonists tested, the pyrin, and linker domains (1-134) function cooperatively and are sufficient for inflammasome activation by certain agonists. Conserved cysteines 8 and 108 appear important for inflammasome activation by sterile, but not infectious insults. Our results define common and agonist-specific regions of NLRP3 that likely mediate ligand-specific responses, discount the hypothesis that NLRP3 inflammasome activation has a unified mechanism, and implicate NLRP3 as an integrator of agonist-specific, inflammasome activating signals.


Sujet(s)
Inflammasomes/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéines adaptatrices de la transduction du signal/immunologie , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Protéines régulatrices de l'apoptose/immunologie , Protéines régulatrices de l'apoptose/métabolisme , Francisella/physiologie , Cellules HEK293 , Humains , Inflammasomes/agonistes , Inflammasomes/composition chimique , Inflammasomes/immunologie , Leucine , Ligands , Listeria monocytogenes/physiologie , Macrophages/immunologie , Macrophages/métabolisme , Souris , Protéine-3 de la famille des NLR contenant un domaine pyrine/agonistes , Protéine-3 de la famille des NLR contenant un domaine pyrine/composition chimique , Protéine-3 de la famille des NLR contenant un domaine pyrine/immunologie , Protéines NLR , Domaine pyrine , Séquences répétées d'acides aminés
4.
J Immunol ; 202(3): 1003-1015, 2019 02 01.
Article de Anglais | MEDLINE | ID: mdl-30598512

RÉSUMÉ

Inflammasome dysregulation is a hallmark of various inflammatory diseases. Evaluating inflammasome-associated structures (ASC specks) and caspase-1 activity by microscopy is time consuming and limited by small sample size. The current flow cytometric method, time of flight inflammasome evaluation (TOFIE), cannot visualize ASC specks or caspase-1 activity, making colocalization studies of inflammasome components and enzymatic activity impossible. We describe a rapid, high-throughput, single-cell, fluorescence-based image analysis method utilizing the Amnis ImageStreamX instrument that quantitatively and qualitatively characterizes the frequency, area, and cellular distribution of ASC specks and caspase-1 activity in mouse and human cells. Unlike TOFIE, this method differentiates between singular perinuclear specks and false positives. With our technique we also show that the presence of NLRP3 reduces the size of ASC specks, which is further reduced by the presence of active caspase-1. The capacity of our approach to simultaneously detect and quantify ASC specks and caspase-1 activity, both at the population and single-cell level, renders it the most powerful tool available for visualizing and quantifying the impact of mutations on inflammasome assembly and activity.


Sujet(s)
Protéines adaptatrices de signalisation CARD/analyse , Caspase-1/analyse , Cytométrie en flux/méthodes , Traitement d'image par ordinateur/méthodes , Inflammasomes/métabolisme , Analyse sur cellule unique/méthodes , Fluorescence , Cellules HEK293 , Humains , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine/agonistes , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Cellules THP-1
5.
Hum Immunol ; 80(1): 32-36, 2019 Jan.
Article de Anglais | MEDLINE | ID: mdl-30056069

RÉSUMÉ

In addition to their role in antigen presentation, class II MHC molecules also transmit signals to B lymphocytes. Class II MHC-mediated signals initiate a range of events in B cells, including induction of cell surface proteins, initiation of cell-cycle progression/proliferation, activation of or protection from apoptosis, and antigen-dependent plasma cell differentiation. Although various transmembrane signaling proteins associate with class II MHC molecules, the class II MHC cytoplasmic domains are essential for signals leading to increased intracellular cAMP and activation of protein kinase C (PKC). Although truncation and mutagenesis studies have provided considerable information about the cytoplasmic domain sequences required, how class II MHC molecules elicit cAMP and PKC activation is not known. Further, appropriate T-dependent B cell responses require intact cAMP and PKC signaling, but the extent to which class II MHC signals are involved is also unknown. This review details our current knowledge of class II MHC cytoplasmic domain signaling in B cells with an emphasis on the likely importance of class II MHC signals for T-dependent antibody responses.


Sujet(s)
Lymphocytes B/métabolisme , Antigènes d'histocompatibilité de classe II/génétique , Antigènes d'histocompatibilité de classe II/métabolisme , Motifs et domaines d'intéraction protéique , Transduction du signal , Séquence d'acides aminés , Animaux , Lymphocytes B/immunologie , Protéines de transport/métabolisme , AMP cyclique , Antigènes d'histocompatibilité de classe II/immunologie , Humains , Liaison aux protéines , Protéine kinase C/métabolisme , Relation structure-activité
6.
J Infect Dis ; 217(9): 1481-1490, 2018 04 11.
Article de Anglais | MEDLINE | ID: mdl-29373737

RÉSUMÉ

Bacterial pneumonia is a common risk factor for acute lung injury and sepsis-mediated death, but the mechanisms underlying the overt inflammation and accompanying pathology are unclear. Infiltration of immature myeloid cells and necrotizing inflammation mediate severe pathology and death during pulmonary infection with Francisella tularensis. However, eliciting mature myeloid cells provides protection. Yet, the host factors responsible for this pathologic immature myeloid cell response are unknown. Here, we report that while the influx of both mature and immature myeloid cells is strictly MyD88 dependent, the interleukin 1 (IL-1) receptor mediates an important dual function via its ligands IL-1α and IL-1ß. Although IL-1ß favors the appearance of bacteria-clearing mature myeloid cells, IL-1α contributes to lung infiltration by ineffective and pathologic immature myeloid cells. Finally, IL-1α and IL-1ß are not the sole factors involved, but myeloid cell responses during acute pneumonia were largely unaffected by lung levels of interleukin 10, interleukin 17, CXCL1, granulocyte colony-stimulating factor, and granulocyte-macrophage colony-stimulating factor.


Sujet(s)
Francisella tularensis , Interleukine-1 alpha/métabolisme , Interleukine-1 bêta/métabolisme , Cellules myéloïdes/physiologie , Tularémie/immunologie , Animaux , Cellules de la moelle osseuse , Femelle , Interleukine-10/génétique , Interleukine-10/métabolisme , Interleukine-17/génétique , Interleukine-17/métabolisme , Interleukine-1 alpha/génétique , Interleukine-1 bêta/génétique , Mâle , Souris , Lignées consanguines de souris , Souris knockout , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/métabolisme , Récepteurs à l'interleukine-1/génétique , Récepteurs à l'interleukine-1/métabolisme , Tularémie/microbiologie , Tularémie/anatomopathologie
7.
Nat Commun ; 8: 15564, 2017 06 05.
Article de Anglais | MEDLINE | ID: mdl-28580947

RÉSUMÉ

Pyrin domain-only proteins (POPs) are recently evolved, primate-specific proteins demonstrated in vitro as negative regulators of inflammatory responses. However, their in vivo function is not understood. Of the four known POPs, only POP2 is reported to regulate NF-κB-dependent transcription and multiple inflammasomes. Here we use a transgenic mouse-expressing POP2 controlled by its endogenous human promotor to study the immunological functions of POP2. Despite having significantly reduced inflammatory cytokine responses to LPS and bacterial infection, POP2 transgenic mice are more resistant to bacterial infection than wild-type mice. In a pulmonary tularaemia model, POP2 enhances IFN-γ production, modulates neutrophil numbers, improves macrophage functions, increases bacterial control and diminishes lung pathology. Thus, unlike other POPs thought to diminish innate protection, POP2 reduces detrimental inflammation while preserving and enhancing protective immunity. Our findings suggest that POP2 acts as a high-order regulator balancing cellular function and inflammation with broad implications for inflammation-associated diseases and therapeutic intervention.


Sujet(s)
Francisella/immunologie , Infections bactériennes à Gram négatif/immunologie , Infections bactériennes à Gram négatif/prévention et contrôle , Inflammation/immunologie , Tularémie/immunologie , Tularémie/prévention et contrôle , Animaux , Lignée cellulaire , Femelle , Cellules HEK293 , Cellules HeLa , Humains , Inflammasomes/immunologie , Interféron gamma/biosynthèse , Interféron gamma/immunologie , Listeria monocytogenes/immunologie , Macrophages/immunologie , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris transgéniques , Protéine-3 de la famille des NLR contenant un domaine pyrine/biosynthèse , Granulocytes neutrophiles/immunologie , Régions promotrices (génétique)/génétique , Salmonella typhimurium/immunologie , Facteur de transcription RelA/biosynthèse , Tularémie/microbiologie , Cellules U937
8.
PLoS Pathog ; 12(12): e1006059, 2016 Dec.
Article de Anglais | MEDLINE | ID: mdl-27926940

RÉSUMÉ

Francisella tularensis (Ft) causes a frequently fatal, acute necrotic pneumonia in humans and animals. Following lethal Ft infection in mice, infiltration of the lungs by predominantly immature myeloid cells and subsequent myeloid cell death drive pathogenesis and host mortality. However, following sub-lethal Ft challenge, more mature myeloid cells are elicited and are protective. In addition, inflammasome-dependent IL-1ß and IL-18 are important for protection. As Nlrp3 appears dispensable for resistance to infection with Francisella novicida, we considered its role during infection with the virulent Type A strain SchuS4 and the attenuated Type B live vaccine strain LVS. Here we show that both in vitro macrophage and in vivo IL-1ß and IL-18 responses to Ft LVS and SchuS4 involve both the Aim2 and Nlrp3 inflammasomes. However, following lethal infection with Francisella, IL-1r-, Caspase-1/11-, Asc- and Aim2-deficient mice exhibited increased susceptibility as expected, while Nlrp3-deficient mice were more resistant. Despite reduced levels of IL-1ß and IL-18, in the absence of Nlrp3, Ft infected mice have dramatically reduced lung pathology, diminished recruitment and death of immature myeloid cells, and reduced bacterial burden in comparison to wildtype and inflammasome-deficient mice. Further, increased numbers of mature neutrophil appear in the lung early during lethal Ft infection in Nlrp3-deficient mice. Finally, Ft infection induces myeloid and lung stromal cell death that in part requires Nlrp3, is necrotic/necroptotic in nature, and drives host mortality. Thus, Nlrp3 mediates an inflammasome-independent process that restricts the appearance of protective mature neutrophils and promotes lethal necrotic lung pathology.


Sujet(s)
Protéine-3 de la famille des NLR contenant un domaine pyrine/immunologie , Granulocytes neutrophiles/immunologie , Pneumopathie bactérienne/immunologie , Tularémie/immunologie , Transfert adoptif , Animaux , Modèles animaux de maladie humaine , Cytométrie en flux , Francisella tularensis/immunologie , Immunohistochimie , Immunophénotypage , Inflammasomes , Souris , Souris de lignée C57BL , Souris transgéniques
9.
J Leukoc Biol ; 100(6): 1335-1347, 2016 12.
Article de Anglais | MEDLINE | ID: mdl-27365531

RÉSUMÉ

IgG (mAb)-opsonized, inactivated Francisella tularensis LVS (iFt-mAb) enhances TLR2-dependent IL-6 production by macrophages via Fcγ receptors (FcγR). In mice, vaccination with iFt-mAb provides IgA-dependent protection against lethal challenge with Ft LVS. Because inflammasome maturation of IL-1ß is thought important for antibody-mediated immunity, we considered the possibility that iFt-mAb elicits an FcγR-dependent myeloid cell inflammasome response. Herein, we find that iFt-mAb enhances macrophage and dendritic cell IL-1ß responses in a TLR2- and FcγR-dependent fashion. Although iFt-mAb complexes bind FcγR and are internalized, sensing of cytosolic DNA by absent in melanoma 2 (AIM2) is not required for the IL-1ß response. In contrast, ASC, caspase-1, and NLR family pyrin domain-containing 3 (NLRP3) are indispensable. Further, FcγR-mediated spleen tyrosine kinase (Syk) signaling is required for this NLRP3-dependent IL-1ß response, but the alternative IL-1ß convertase caspase-8 is insufficient. Finally, iFt-mAb-vaccinated wild-type mice exhibit a significant delay in time to death, but IL-1R1- or Nlrp3-deficient mice vaccinated in this way are not protected and lack appreciable Francisella-specific antibodies. This study demonstrates that FcγR-mediated Syk activation leads to NLRP3 inflammasome-dependent IL-1ß production in macrophages and suggests that an Nlrp3- and IL-1R-dependent process contributes to the IgA response important for protection against Ft LVS. These findings extend our understanding of cellular responses to inactivated pathogen-opsonized vaccine, establish FcγR-elicited Syk kinase-mediated NLRP3 inflammasome activation, and provide additional insight toward understanding crosstalk between TLR and FcγR signals.


Sujet(s)
Anticorps antibactériens/immunologie , Complexe antigène-anticorps/immunologie , Antigènes bactériens/immunologie , Francisella tularensis/immunologie , Inflammasomes/immunologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/immunologie , Récepteurs du fragment Fc des IgG/immunologie , Syk kinase/immunologie , Récepteur de type Toll-2/immunologie , Animaux , Anticorps monoclonaux/immunologie , Vaccins antibactériens , Caspase 8/immunologie , Lignée cellulaire , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Interleukine-1 bêta/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Souris de lignée C57BL , Souris knockout , Monocytes/immunologie , Monocytes/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/déficit , Transduction du signal , Vaccination
10.
PLoS Pathog ; 12(3): e1005517, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-27015566

RÉSUMÉ

Inhalation of Francisella tularensis (Ft) causes acute and fatal pneumonia. The lung cytokine milieu favors exponential Ft replication, but the mechanisms underlying acute pathogenesis and death remain unknown. Evaluation of the sequential and systemic host immune response in pulmonary tularemia reveals that in contrast to overwhelming bacterial burden or cytokine production, an overt innate cellular response to Ft drives tissue pathology and host mortality. Lethal infection with Ft elicits medullary and extra-medullary myelopoiesis supporting recruitment of large numbers of immature myeloid cells and MDSC to the lungs. These cells fail to mature and die, leading to subsequent necrotic lung damage, loss of pulmonary function, and host death that is partially dependent upon immature Ly6G+ cells. Acceleration of this process may account for the rapid lethality seen with Ft SchuS4. In contrast, during sub-lethal infection with Ft LVS the pulmonary cellular response is characterized by a predominance of mature neutrophils and monocytes required for protection, suggesting a required threshold for lethal bacterial infection. Further, eliciting a mature phagocyte response provides transient, but dramatic, innate protection against Ft SchuS4. This study reveals that the nature of the myeloid cell response may be the primary determinant of host mortality versus survival following Francisella infection.


Sujet(s)
Francisella tularensis/immunologie , Récepteur de type Toll-2/métabolisme , Tularémie/immunologie , Animaux , Cytokines/métabolisme , Humains , Inflammation , Poumon/immunologie , Souris de lignée C57BL , Cellules myéloïdes/métabolisme , Pneumopathie infectieuse/métabolisme
12.
J Biol Chem ; 289(17): 11695-11703, 2014 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-24619409

RÉSUMÉ

Major histocompatibility complex (MHC) class II molecules exhibit conformational heterogeneity, which influences their ability to stimulate CD4 T cells and drive immune responses. Previous studies suggest a role for the transmembrane domain of the class II αß heterodimer in determining molecular structure and function. Our previous studies identified an MHC class II conformer that is marked by the Ia.2 epitope. These Ia.2(+) class II conformers are lipid raft-associated and able to drive both tyrosine kinase signaling and efficient antigen presentation to CD4 T cells. Here, we establish that the Ia.2(+) I-A(k) conformer is formed early in the class II biosynthetic pathway and that differential pairing of highly conserved transmembrane domain GXXXG dimerization motifs is responsible for formation of Ia.2(+) versus Ia.2(-) I-A(k) class II conformers and controlling lipid raft partitioning. These findings provide a molecular explanation for the formation of two distinct MHC class II conformers that differ in their inherent ability to signal and drive robust T cell activation, providing new insight into the role of MHC class II in regulating antigen-presenting cell-T cell interactions critical to the initiation and control of multiple aspects of the immune response.


Sujet(s)
Antigènes d'histocompatibilité de classe II/immunologie , Motifs d'acides aminés , Séquence d'acides aminés , Lignée cellulaire , Dimérisation , Réticulum endoplasmique/immunologie , Antigènes d'histocompatibilité de classe II/composition chimique , Données de séquences moléculaires , Structure moléculaire , Similitude de séquences d'acides aminés
13.
J Immunol ; 192(4): 1946-53, 2014 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-24446520

RÉSUMÉ

Using several tumor models, we demonstrate that mice deficient in Bcl11b in T cells, although having reduced numbers of T cells in the peripheral lymphoid organs, developed significantly less tumors compared with wild-type mice. Bcl11b(-/-) CD4(+) T cells, with elevated TNF-α levels, but not the Bcl11b(-/-) CD8(+) T cells, were required for the reduced tumor burden, as were NK1.1(+) cells, found in increased numbers in Bcl11b(F/F)/CD4-Cre mice. Among NK1.1(+) cells, the NK cell population was predominant in number and was the only population displaying elevated granzyme B levels and increased degranulation, although not increased proliferation. Although the number of myeloid-derived suppressor cells was increased in the lungs with metastatic tumors of Bcl11b(F/F)/CD4-Cre mice, their arginase-1 levels were severely reduced. The increase in NK cell and myeloid-derived suppressor cell numbers was associated with increased bone marrow and splenic hematopoiesis. Finally, the reduced tumor burden, increased numbers of NK cells in the lung, and increased hematopoiesis in Bcl11b(F/F)/CD4-Cre mice were all dependent on TNF-α. Moreover, TNF-α treatment of wild-type mice also reduced the tumor burden and increased hematopoiesis and the numbers and activity of NK cells in the lung. In vitro treatment with TNF-α of lineage-negative hematopoietic progenitors increased NK and myeloid differentiation, further supporting a role of TNF-α in promoting hematopoiesis. These studies reveal a novel role for TNF-α in the antitumor immune response, specifically in stimulating hematopoiesis and increasing the numbers and activity of NK cells.


Sujet(s)
Mélanome/immunologie , Mélanome/anatomopathologie , Protéines de répression/métabolisme , Lymphocytes T régulateurs/immunologie , Charge tumorale/immunologie , Facteur de nécrose tumorale alpha/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Animaux , Antigènes Ly/métabolisme , Arginase/métabolisme , Lymphocytes T CD8+/immunologie , Dégranulation cellulaire/immunologie , Prolifération cellulaire , Délétion de gène , Granzymes/métabolisme , Hématopoïèse , Cellules tueuses naturelles/immunologie , Tumeurs du poumon/immunologie , Numération des lymphocytes , Souris , Souris transgéniques , Cellules myéloïdes/immunologie , Sous-famille B des récepteurs de cellules NK de type lectine/métabolisme , Protéines de répression/génétique , Rate/immunologie , Lymphocytes T régulateurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Régulation positive
14.
Front Immunol ; 4: 275, 2013 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-24062743

RÉSUMÉ

Upon activation Nod-like receptors (NLRs) assemble into multi-protein complexes such as the NODosome and inflammasome. This process relies upon homo domain interactions between the structurally related Pyrin and caspase-recruitment (CARD) domains and adaptor proteins, such as ASC, or effector proteins, such as caspase-1. Although a variety of NLRP and NLRC complexes have been described along with their activating stimuli and associated proteins, less familiar are processes limiting assembly and/or promoting dissociation of NLR complexes. Given the importance of limiting harmful, chronic inflammation, such regulatory mechanisms are significant and likely numerous. Proteins comprised of a solitary Pyrin domain (Pyrin-only) or CARD domain (CARD-only) posses an obvious potential ability to act as competitive inhibitors of NLR complexes. Indeed, both Pyrin-only proteins (POPs) and CARD-only proteins (COPs) have been described as regulators of caspase-1 and/or NLR-inflammasome activation and not surprisingly as factors mediating pathogenesis. Although clear examples of pathogen encoded POPs are currently limited to members of the poxviridae, the human genome likely encodes three POPs (POP1, POP2, and a potential POP3), of which only POP2 is known to prevent NLR:ASC interaction, and three COPs (COP/Pseudo-ICE, INCA, and ICEBERG), initially described for their ability to inhibit caspase-1 activity. Surprisingly, among eukaryotic species POPs and COPs appear to be evolutionarily recent and restricted to higher primates, suggesting strong selective pressures driving their emergence. Despite the importance of understanding the regulation of NLR functions, relatively little attention has been devoted to revealing the biological impact of these intriguing proteins. This review highlights the current state of our understanding of POPs and COPs with attention to protein interaction, functions, evolution, implications for health and disease, and outstanding questions.

15.
J Biol Chem ; 288(33): 23844-57, 2013 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-23821549

RÉSUMÉ

Francisella tularensis is an important human pathogen responsible for causing tularemia. F. tularensis has long been developed as a biological weapon and is now classified as a category A agent by the Centers for Disease Control because of its possible use as a bioterror agent. F. tularensis represses inflammasome; a cytosolic multi-protein complex that activates caspase-1 to produce proinflammatory cytokines IL-1ß and IL-18. However, the Francisella factors and the mechanisms through which F. tularensis mediates these suppressive effects remain relatively unknown. Utilizing a mutant of F. tularensis in FTL_0325 gene, this study investigated the mechanisms of inflammasome repression by F. tularensis. We demonstrate that muted IL-1ß and IL-18 responses generated in macrophages infected with F. tularensis live vaccine strain (LVS) or the virulent SchuS4 strain are due to a predominant suppressive effect on TLR2-dependent signal 1. Our results also demonstrate that FTL_0325 of F. tularensis impacts proIL-1ß expression as early as 2 h post-infection and delays activation of AIM2 and NLRP3-inflammasomes in a TLR2-dependent fashion. An enhanced activation of caspase-1 and IL-1ß observed in FTL_0325 mutant-infected macrophages at 24 h post-infection was independent of both AIM2 and NLRP3. Furthermore, F. tularensis LVS delayed pyroptotic cell death of the infected macrophages in an FTL_0325-dependent manner during the early stages of infection. In vivo studies in mice revealed that suppression of IL-1ß by FTL_0325 early during infection facilitates the establishment of a fulminate infection by F. tularensis. Collectively, this study provides evidence that F. tularensis LVS represses inflammasome activation and that F. tularensis-encoded FTL_0325 mediates this effect.


Sujet(s)
Francisella tularensis/immunologie , Inflammasomes/métabolisme , Tularémie/immunologie , Tularémie/microbiologie , Animaux , Protéines de transport/métabolisme , Mort cellulaire , Protéines de liaison à l'ADN , Humains , Interféron bêta/métabolisme , Interleukine-18/métabolisme , Interleukine-1 bêta/métabolisme , Macrophages/métabolisme , Macrophages/microbiologie , Macrophages/anatomopathologie , Souris , Souris de lignée C57BL , Mutation/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine , Protéines nucléaires/métabolisme , Transduction du signal/immunologie , Récepteur de type Toll-2/métabolisme , Récepteur de type Toll-4/métabolisme
16.
J Biol Chem ; 287(30): 25216-29, 2012 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-22654100

RÉSUMÉ

Francisella tularensis, the causative agent of tularemia, is one of the deadliest agents of biological warfare and bioterrorism. Extremely high virulence of this bacterium is associated with its ability to dampen or subvert host innate immune response. The objectives of this study were to identify factors and understand the mechanisms of host innate immune evasion by F. tularensis. We identified and explored the pathogenic role of a mutant interrupted at gene locus FTL_0325, which encodes an OmpA-like protein. Our results establish a pathogenic role of FTL_0325 and its ortholog FTT0831c in the virulent F. tularensis SchuS4 strain in intramacrophage survival and suppression of proinflammatory cytokine responses. This study provides mechanistic evidence that the suppressive effects on innate immune responses are due specifically to these proteins and that FTL_0325 and FTT0831c mediate immune subversion by interfering with NF-κB signaling. Furthermore, FTT0831c inhibits NF-κB activity primarily by preventing the nuclear translocation of p65 subunit. Collectively, this study reports a novel F. tularensis factor that is required for innate immune subversion caused by this deadly bacterium.


Sujet(s)
Protéines de la membrane externe bactérienne/immunologie , Francisella tularensis/immunologie , Francisella tularensis/pathogénicité , Immunité innée , Macrophages/immunologie , Tularémie/immunologie , Facteurs de virulence/immunologie , Animaux , Protéines de la membrane externe bactérienne/génétique , Lignée cellulaire , Francisella tularensis/génétique , Locus génétiques/immunologie , Humains , Macrophages/microbiologie , Souris , Souris de lignée BALB C , Transduction du signal/génétique , Transduction du signal/immunologie , Facteur de transcription RelA/génétique , Facteur de transcription RelA/immunologie , Tularémie/génétique , Facteurs de virulence/génétique
17.
PLoS One ; 7(5): e37330, 2012.
Article de Anglais | MEDLINE | ID: mdl-22615981

RÉSUMÉ

Francisella tularensis is a bacterial pathogen that uses host-derived PGE2 to subvert the host's adaptive immune responses in multiple ways. Francisella-induced PGE2 acts directly on CD4 T cells to blunt production of IFN-γ. Francisella-induced PGE2 can also elicit production of a >10 kDa soluble host factor termed FTMØSN (F. tularensismacrophage supernatant), which acts on IFN-γ pre-activated MØ to down-regulate MHC class II expression via a ubiquitin-dependent mechanism, blocking antigen presentation to CD4 T cells. Here, we report that FTMØSN-induced down-regulation of MØ class II is the result of the induction of MARCH1, and that MØ expressing MARCH1 "resistant" class II molecules are resistant to FTMØSN-induced class II down-regulation. Since PGE2 can induce IL-10 production and IL-10 is the only reported cytokine able to induce MARCH1 expression in monocytes and dendritic cells, these findings suggested that IL-10 is the active factor in FTMØSN. However, use of IL-10 knockout MØ established that IL-10 is not the active factor in FTMØSN, but rather that Francisella-elicited PGE2 drives production of a >10 kDa host factor distinct from IL-10. This factor then drives MØ IL-10 production to induce MARCH1 expression and the resultant class II down-regulation. Since many human pathogens such as Salmonella typhi, Mycobacterium tuberculosis and Legionella pneumophila also induce production of host PGE2, these results suggest that a yet-to-be-identified PGE2-inducible host factor capable of inducing IL-10 is central to the immune evasion mechanisms of multiple important human pathogens.


Sujet(s)
Lymphocytes T CD4+/immunologie , Francisella tularensis/immunologie , Antigènes d'histocompatibilité de classe II/biosynthèse , Interleukine-10/biosynthèse , Macrophages/immunologie , Ubiquitin-protein ligases/biosynthèse , Dinoprostone/physiologie , Régulation négative , Humains , Interleukine-10/génétique , Macrophages/métabolisme , Ubiquitin-protein ligases/physiologie , Régulation positive
18.
J Biol Chem ; 286(47): 40536-47, 2011 Nov 25.
Article de Anglais | MEDLINE | ID: mdl-21976665

RÉSUMÉ

Activation of transcription factor NF-κB and inflammasome-directed caspase-1 cleavage of IL-1ß are key processes in the inflammatory response to pathogen or host-derived signals. Pyrin-only proteins (POPs) are restricted to Old World monkeys, apes, and humans and have previously been shown to impair inflammasome assembly and/or NF-κB p65 transcriptional activity in transfected epithelial cells. However, the biological role of POP2 and the molecular basis for its observed functions are not well understood. In this report we demonstrate that POP2 regulates TNFα and IL-1ß responses in human monocytic THP-1 cells and in stable transfectants of mouse J774A.1 macrophages. Deletion analysis of POP2 revealed that the first α-helix (residues 1-19) is necessary and sufficient for both inflammasome and NF-κB inhibitory functions. Further, key acidic residues Glu(6), Asp(8), and Glu(16), believed critical for Pyrin/Pyrin domain interaction, are important for inflammasome inhibition. Moreover, these mutations did not reduce the effect of POP2 upon NF-κB, indicating that the inflammasome and NF-κB inhibitory properties of POP2 can be uncoupled mechanistically. Collectively, these data demonstrate that POP2 acts as a regulator of inflammatory signals and exerts its two known functions through distinct modalities employed by its first α-helix.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Inflammasomes/métabolisme , Facteur de transcription RelA/métabolisme , Protéines adaptatrices de la transduction du signal/composition chimique , Protéines adaptatrices de la transduction du signal/génétique , Séquence d'acides aminés , Animaux , Protéines régulatrices de l'apoptose , Protéines adaptatrices de signalisation CARD , Protéines de transport/métabolisme , Protéines du cytosquelette/métabolisme , Francisella/physiologie , Cellules HEK293 , Humains , Interleukine-1 bêta/métabolisme , Lipopolysaccharides/pharmacologie , Macrophages/cytologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/microbiologie , Souris , Données de séquences moléculaires , Monocytes/cytologie , Monocytes/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Mutagenèse , Mutation , Protéine-3 de la famille des NLR contenant un domaine pyrine , Structure secondaire des protéines , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription RelA/génétique , Activation de la transcription/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme
19.
J Biol Chem ; 286(45): 39033-42, 2011 Nov 11.
Article de Anglais | MEDLINE | ID: mdl-21930705

RÉSUMÉ

Pathogen-triggered activation of the inflammasome complex leading to caspase-1 activation and IL-1ß production involves similar sensor proteins between mouse and human. However, the specific sensors used may differ between infectious agents and host species. In mice, Francisella infection leads to seemingly exclusive activation of the Aim2 inflammasome with no apparent role for Nlrp3. Here we examine the IL-1ß response of human cells to Francisella infection. Francisella strains exhibit differences in IL-1ß production by influencing induction of IL-1ß and ASC transcripts. Unexpectedly, our results demonstrate that Francisella activates the NLRP3 inflammasome in human cells. Francisella infection of THP-1 cells elicits IL-1ß production, which is reduced by siRNA targeting of NLRP3. Moreover, in reconstituted 293T cells, Francisella triggers assembly of the NLRP3 inflammasome complex. In addition, inhibitors of reactive oxygen species, cathepsin B, and K(+) efflux pathways, known to specifically influence NLRP3, substantially but not completely impair the Francisella-elicited IL-1ß response, suggesting the involvement of another inflammasome pathway. Finally, shRNA targeting of NLRP3 and AIM2 reveals that both pathways contribute to the inflammasome response. Together these results establish NLRP3 as a cytosolic sensor for Francisella in human cells, a role not observed in mouse.


Sujet(s)
Protéines de transport/métabolisme , Francisella tularensis/métabolisme , Inflammasomes/métabolisme , Tularémie/métabolisme , Animaux , Protéines de transport/génétique , Cathepsine B/génétique , Cathepsine B/métabolisme , Protéines de liaison à l'ADN , Cellules HEK293 , Humains , Inflammasomes/génétique , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Transport des ions/génétique , Souris , Protéine-3 de la famille des NLR contenant un domaine pyrine , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Potassium/métabolisme , Espèces réactives de l'oxygène/métabolisme , Spécificité d'espèce , Tularémie/génétique
20.
J Immunol ; 186(12): 6710-7, 2011 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-21543648

RÉSUMÉ

Previous work established that binding of the 11-5.2 anti-I-A(k) mAb, which recognizes the Ia.2 epitope on I-A(k) class II molecules, elicits MHC class II signaling, whereas binding of two other anti-I-A(k) mAbs that recognize the Ia.17 epitope fail to elicit signaling. Using a biochemical approach, we establish that the Ia.2 epitope recognized by the widely used 11-5.2 mAb defines a subset of cell surface I-A(k) molecules predominantly found within membrane lipid rafts. Functional studies demonstrate that the Ia.2-bearing subset of I-A(k) class II molecules is critically necessary for effective B cell-T cell interactions, especially at low Ag doses, a finding consistent with published studies on the role of raft-resident class II molecules in CD4 T cell activation. Interestingly, B cells expressing recombinant I-A(k) class II molecules possessing a ß-chain-tethered hen egg lysosome peptide lack the Ia.2 epitope and fail to partition into lipid rafts. Moreover, cells expressing Ia.2(-) tethered peptide-class II molecules are severely impaired in their ability to present both tethered peptide or peptide derived from exogenous Ag to CD4 T cells. These results establish the Ia.2 epitope as defining a lipid raft-resident MHC class II conformer vital to the initiation of MHC class II-restricted B cell-T cell interactions.


Sujet(s)
Présentation d'antigène/immunologie , Épitopes/immunologie , Antigènes d'histocompatibilité de classe II/immunologie , Microdomaines membranaires/immunologie , Animaux , Lymphocytes B/immunologie , Communication cellulaire/immunologie , Humains , Activation des lymphocytes , Microdomaines membranaires/composition chimique , Souris , Lymphocytes T/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE