Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 2193, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366851

ABSTRACT

Innate immunity to nucleic acids forms the backbone for anti-viral immunity and several inflammatory diseases. Upon sensing cytosolic viral RNA, retinoic acid-inducible gene-I-like receptors (RLRs) interact with the mitochondrial antiviral signaling protein (MAVS) and activate TANK-binding kinase 1 (TBK1) to induce type I interferon (IFN-I). TRAF3-interacting protein 3 (TRAF3IP3, T3JAM) is essential for T and B cell development. It is also well-expressed by myeloid cells, where its role is unknown. Here we report that TRAF3IP3 suppresses cytosolic poly(I:C), 5'ppp-dsRNA, and vesicular stomatitis virus (VSV) triggers IFN-I expression in overexpression systems and Traf3ip3-/- primary myeloid cells. The mechanism of action is through the interaction of TRAF3IP3 with endogenous TRAF3 and TBK1. This leads to the degradative K48 ubiquitination of TBK1 via its K372 residue in a DTX4-dependent fashion. Mice with myeloid-specific gene deletion of Traf3ip3 have increased RNA virus-triggered IFN-I production and reduced susceptibility to virus. These results identify a function of TRAF3IP3 in the regulation of the host response to cytosolic viral RNA in myeloid cells.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation , Interferon Type I/genetics , Membrane Proteins/genetics , Myeloid Cells/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Viral/genetics , Animals , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Chlorocebus aethiops , Cytosol/metabolism , Cytosol/virology , HEK293 Cells , HeLa Cells , Humans , Interferon Type I/metabolism , Jurkat Cells , Lysine/genetics , Lysine/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/virology , Protein Serine-Threonine Kinases/metabolism , RNA, Viral/metabolism , THP-1 Cells , Ubiquitination , Vero Cells , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/physiology
2.
Methods Mol Biol ; 1960: 41-55, 2019.
Article in English | MEDLINE | ID: mdl-30798519

ABSTRACT

Macrophages are cellular components of the immune system that are essential for responding to pathogens, initiating inflammation and maintaining tissue homeostasis. Isolation, culture, and functional characterization of bone marrow-derived macrophages from mice are exceptionally powerful in vitro techniques used to examine aspects of macrophage biology, including effector functions, such as phagocytosis, cytokine secretion, oxidative burst, migration, and antigen processing and presentation. These studies can be carried out using wild-type, gene-ablated, and/or transgenic mice. The quantity, purity, and ease of culture of these cells enhance their utility for primary cell cultures to understand macrophage biology. Mouse macrophages have become a cognate animal model for the study of human macrophage biology and disease. This chapter outlines protocols used to generate, polarize, quantitate, and functionally evaluate macrophages derived from bone marrow precursor cells.


Subject(s)
Bone Marrow Cells/cytology , Macrophages/cytology , Animals , Bone Marrow Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Inflammation/immunology , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Transgenic , Phagocytosis/physiology , Respiratory Burst/physiology
3.
J Immunol ; 199(8): 2896-2909, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28864474

ABSTRACT

Sensing of cytosolic nucleotides is a critical initial step in the elaboration of type I IFN. One of several upstream receptors, cyclic GMP-AMP synthase, binds to cytosolic DNA and generates dicyclic nucleotides that act as secondary messengers. These secondary messengers bind directly to stimulator of IFN genes (STING). STING recruits TNFR-associated NF-κB kinase-binding kinase 1 which acts as a critical node that allows for efficient activation of IFN regulatory factors to drive the antiviral transcriptome. NLRC3 is a recently characterized nucleotide-binding domain, leucine-rich repeat containing protein (NLR) that negatively regulates the type I IFN pathway by inhibiting subcellular redistribution and effective signaling of STING, thus blunting the transcription of type I IFNs. NLRC3 is predominantly expressed in lymphoid and myeloid cells. IQGAP1 was identified as a putative interacting partner of NLRC3 through yeast two-hybrid screening. In this article, we show that IQGAP1 associates with NLRC3 and can disrupt the NLRC3-STING interaction in the cytosol of human epithelial cells. Furthermore, knockdown of IQGAP1 in THP1 and HeLa cells causes significantly more IFN-ß production in response to cytosolic nucleic acids. This result phenocopies NLRC3-deficient macrophages and fibroblasts and short hairpin RNA knockdown of NLRC3 in THP1 cells. Our findings suggest that IQGAP1 is a novel regulator of type I IFN production, possibly via interacting with NLRC3 in human monocytic and epithelial cells.


Subject(s)
Epithelial Cells/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/physiology , Virus Diseases/immunology , ras GTPase-Activating Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunity , Interferon Type I/metabolism , Membrane Proteins/metabolism , Nucleic Acids/immunology , Protein Binding , RNA, Small Interfering/genetics , Signal Transduction
4.
J Biol Chem ; 291(24): 12747-12760, 2016 Jun 10.
Article in English | MEDLINE | ID: mdl-27068743

ABSTRACT

Most colon cancer cases are initiated by truncating mutations in the tumor suppressor, adenomatous polyposis coli (APC). APC is a critical negative regulator of the Wnt signaling pathway that participates in a multi-protein "destruction complex" to target the key effector protein ß-catenin for ubiquitin-mediated proteolysis. Prior work has established that the poly(ADP-ribose) polymerase (PARP) enzyme Tankyrase (TNKS) antagonizes destruction complex activity by promoting degradation of the scaffold protein Axin, and recent work suggests that TNKS inhibition is a promising cancer therapy. We performed a yeast two-hybrid (Y2H) screen and uncovered TNKS as a putative binding partner of Drosophila APC2, suggesting that TNKS may play multiple roles in destruction complex regulation. We find that TNKS binds a C-terminal RPQPSG motif in Drosophila APC2, and that this motif is conserved in human APC2, but not human APC1. In addition, we find that APC2 can recruit TNKS into the ß-catenin destruction complex, placing the APC2/TNKS interaction at the correct intracellular location to regulate ß-catenin proteolysis. We further show that TNKS directly PARylates both Drosophila Axin and APC2, but that PARylation does not globally regulate APC2 protein levels as it does for Axin. Moreover, TNKS inhibition in colon cancer cells decreases ß-catenin signaling, which we find cannot be explained solely through Axin stabilization. Instead, our findings suggest that TNKS regulates destruction complex activity at the level of both Axin and APC2, providing further mechanistic insight into TNKS inhibition as a potential Wnt pathway cancer therapy.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Axin Protein/metabolism , Cytoskeletal Proteins/metabolism , Tankyrases/metabolism , beta Catenin/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Axin Protein/genetics , Blotting, Western , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Fluorescent Antibody Technique , HCT116 Cells , Humans , Male , Protein Binding , Substrate Specificity , Tankyrases/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Two-Hybrid System Techniques , beta Catenin/genetics
5.
Nat Med ; 21(8): 906-13, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26107252

ABSTRACT

The inflammasome activates caspase-1 and the release of interleukin-1ß (IL-1ß) and IL-18, and several inflammasomes protect against intestinal inflammation and colitis-associated colon cancer (CAC) in animal models. The absent in melanoma 2 (AIM2) inflammasome is activated by double-stranded DNA, and AIM2 expression is reduced in several types of cancer, but the mechanism by which AIM2 restricts tumor growth remains unclear. We found that Aim2-deficient mice had greater tumor load than Asc-deficient mice in the azoxymethane/dextran sodium sulfate (AOM/DSS) model of colorectal cancer. Tumor burden was also higher in Aim2(-/-)/Apc(Min/+) than in APC(Min/+) mice. The effects of AIM2 on CAC were independent of inflammasome activation and IL-1ß and were primarily mediated by a non-bone marrow source of AIM2. In resting cells, AIM2 physically interacted with and limited activation of DNA-dependent protein kinase (DNA-PK), a PI3K-related family member that promotes Akt phosphorylation, whereas loss of AIM2 promoted DNA-PK-mediated Akt activation. AIM2 reduced Akt activation and tumor burden in colorectal cancer models, while an Akt inhibitor reduced tumor load in Aim2(-/-) mice. These findings suggest that Akt inhibitors could be used to treat AIM2-deficient human cancers.


Subject(s)
Colonic Neoplasms/prevention & control , DNA-Activated Protein Kinase/physiology , DNA-Binding Proteins/physiology , Inflammasomes/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Colitis/complications , Female , HCT116 Cells , Humans , Intestinal Polyps/prevention & control , Male , Mice , Mice, Inbred C57BL , Phosphorylation
6.
Inflamm Bowel Dis ; 20(12): 2412-32, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25153506

ABSTRACT

Pattern recognition receptors are essential mediators of host defense and inflammation in the gastrointestinal system. Recent data have revealed that toll-like receptors and nucleotide-binding domain and leucine-rich repeat-containing proteins (NLRs) function to maintain homeostasis between the host microbiome and mucosal immunity. The NLR proteins are a diverse class of cytoplasmic pattern recognition receptors. In humans, only about half of the identified NLRs have been adequately characterized. The majority of well-characterized NLRs participate in the formation of a multiprotein complex, termed the inflammasome, which is responsible for the maturation of interleukin-1ß and interleukin-18. However, recent observations have also uncovered the presence of a novel subgroup of NLRs that function as positive or negative regulators of inflammation through modulating critical signaling pathways, including NF-κB. Dysregulation of specific NLRs from both proinflammatory and inhibitory subgroups have been associated with the development of inflammatory bowel disease (IBD) in genetically susceptible human populations. Our own preliminary retrospective data mining efforts have identified a diverse range of NLRs that are significantly altered at the messenger RNA level in colons from patients with IBD. Likewise, studies using genetically modified mouse strains have revealed that multiple NLR family members have the potential to dramatically modulate the immune response during IBD. Targeting NLR signaling represents a promising and novel therapeutic strategy. However, significant effort is necessary to translate the current understanding of NLR biology into effective therapies.


Subject(s)
Inflammatory Bowel Diseases/physiopathology , Receptors, Cell Surface/metabolism , Animals , Humans , Mice
7.
Methods Mol Biol ; 1032: 79-89, 2013.
Article in English | MEDLINE | ID: mdl-23943445

ABSTRACT

The role of macrophage subsets in allergic diseases in vivo is under current investigation. These cells perform sentinel functions in the lung, the skin, and the gastrointestinal mucosa. Their interface with environmental cues influences the initiation, progression, development, and resolution of allergic diseases. Researchers often culture bone marrow-derived macrophages to study macrophage biology. The in vitro maturation of bone marrow precursor cells into mature macrophages is a powerful technique used to study macrophage biology. The polarization or differential activation of macrophages into functionally distinct subsets can provide insight into allergic disease pathologies. Classically activated, alternatively activated, and regulatory macrophages have different effector functions that can affect allergic responses. Understanding macrophage biology during allergen exposure, host sensitization, and disease progression/resolution may lead to improved therapeutic interventions. The purpose of this chapter is to outline protocols used for the culture and polarization of classically activated, alternatively activated, and regulatory macrophages. In addition, the techniques to measure macrophage-specific effector molecules by ELISA, RT-PCR, and immunoblotting are reviewed.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Macrophage Activation , Macrophages/metabolism , Animals , Bone Marrow Cells/metabolism , Cytokines/metabolism , Humans , Macrophages/cytology , Mice
8.
Methods Mol Biol ; 1031: 27-35, 2013.
Article in English | MEDLINE | ID: mdl-23824883

ABSTRACT

Macrophages are cellular components of the immune system that are essential for responding to pathogens, initiating inflammation, and maintaining tissue homeostasis. Isolation, culture, and functional characterization of bone marrow-derived macrophages from mice are exceptionally powerful techniques used to examine aspects of macrophage biology in vitro. These cells can be used to study effector functions, such as phagocytosis, cytokine secretion, oxidative burst, migration, antigen processing and presentation, in the context of wild-type, gene-ablated, and/or transgenic mice. The quantity, purity, and ease of culture of these cells enhance their utility for primary cell cultures. This chapter outlines protocols used to generate, quantitate, and functionally evaluate macrophages derived from bone marrow precursor cells.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Macrophages/cytology , Animals , Mice , Mice, Transgenic/genetics
9.
PLoS Pathog ; 9(4): e1003256, 2013.
Article in English | MEDLINE | ID: mdl-23592984

ABSTRACT

Influenza A virus (IAV) triggers a contagious and potentially lethal respiratory disease. A protective IL-1ß response is mediated by innate receptors in macrophages and lung epithelial cells. NLRP3 is crucial in macrophages; however, which sensors elicit IL-1ß secretion in lung epithelial cells remains undetermined. Here, we describe for the first time the relative roles of the host innate receptors RIG-I (DDX58), TLR3, and NLRP3 in the IL-1ß response to IAV in primary lung epithelial cells. To activate IL-1ß secretion, these cells employ partially redundant recognition mechanisms that differ from those described in macrophages. RIG-I had the strongest effect through a MAVS/TRIM25/Riplet-dependent type I IFN signaling pathway upstream of TLR3 and NLRP3. Notably, RIG-I also activated the inflammasome through interaction with caspase 1 and ASC in primary lung epithelial cells. Thus, NS1, an influenza virulence factor that inhibits the RIG-I/type I IFN pathway, strongly modulated the IL-1ß response in lung epithelial cells and in ferrets. The NS1 protein derived from a highly pathogenic strain resulted in increased interaction with RIG-I and inhibited type I IFN and IL-1ß responses compared to the least pathogenic virus strains. These findings demonstrate that in IAV-infected lung epithelial cells RIG-I activates the inflammasome both directly and through a type I IFN positive feedback loop.


Subject(s)
Carrier Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Inflammasomes/metabolism , Influenza A Virus, H1N1 Subtype , Interferon-beta/metabolism , Toll-Like Receptor 3/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , Cells, Cultured , Cytoskeletal Proteins/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Epithelial Cells/metabolism , Ferrets , HEK293 Cells , Humans , Influenza A Virus, H1N1 Subtype/metabolism , Lung/metabolism , Lung/virology , Macrophages/immunology , Male , NLR Family, Pyrin Domain-Containing 3 Protein , RNA Interference , Receptors, Immunologic , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Signal Transduction , Toll-Like Receptor 3/genetics , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism , Viral Nonstructural Proteins/metabolism
10.
Nat Immunol ; 13(9): 823-31, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22863753

ABSTRACT

Several members of the NLR family of sensors activate innate immunity. In contrast, we found here that NLRC3 inhibited Toll-like receptor (TLR)-dependent activation of the transcription factor NF-κB by interacting with the TLR signaling adaptor TRAF6 to attenuate Lys63 (K63)-linked ubiquitination of TRAF6 and activation of NF-κB. We used bioinformatics to predict interactions between NLR and TRAF proteins, including interactions of TRAF with NLRC3. In vivo, macrophage expression of Nlrc3 mRNA was diminished by the administration of lipopolysaccharide (LPS) but was restored when cellular activation subsided. To assess biologic relevance, we generated Nlrc3(-/-) mice. LPS-treated Nlrc3(-/-) macrophages had more K63-ubiquitinated TRAF6, nuclear NF-κB and proinflammatory cytokines. Finally, LPS-treated Nlrc3(-/-) mice had more signs of inflammation. Thus, signaling via NLRC3 and TLR constitutes a negative feedback loop. Furthermore, prevalent NLR-TRAF interactions suggest the formation of a 'TRAFasome' complex.


Subject(s)
NF-kappa B/immunology , Receptors, G-Protein-Coupled/immunology , Signal Transduction/immunology , TNF Receptor-Associated Factor 6/immunology , Toll-Like Receptors/immunology , Amino Acid Sequence , Animals , Feedback, Physiological , HEK293 Cells , Humans , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , NF-kappa B/metabolism , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptors/metabolism
11.
J Biol Chem ; 287(29): 24294-303, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22645137

ABSTRACT

Most of the nucleotide-binding domain, leucine-rich repeat (NLR) proteins regulate responses to microbial and damage-associated products. Class II transactivator (CIITA) has a distinct function as the master regulator of class II major histocompatibility complex (MHC-II) transcription. Recently, human NLRC5 was found to regulate MHC-I in cell lines; however, a host of conflicting positive and negative functions has been attributed to this protein. To address the function of NLRC5 in a physiologic setting, we generated an Nlrc5(-/-) strain that contains a deletion in the exon that encodes the nucleotide-binding domain. We have not detected a role for this protein in cytokine induction by pathogen-associated molecular patterns and viruses. However, Nlrc5(-/-) cells showed a dramatic decrease of classical (H-2K) and nonclassical (Tla) MHC-I expression by T/B lymphocytes, natural killer (NK) cells, and myeloid-monocytic lineages. As a comparison, CIITA did not affect mouse MHC-I expression. Nlrc5(-/-) splenocytes and bone marrow-derived macrophages were able to up-regulate MHC-I in response to IFN-γ; however, the absolute levels of MHC-I expression were significantly lower than WT controls. Chromatin immunoprecipitation of IFN-γ-treated cells indicates that Nlrc5 reduced the silencing H3K27me3 histone modification, but did not affect the activating AcH3 modification on a MHC-I promoter. In summary, we conclude that Nlrc5 is important in the regulation of MHC-I expression by reducing H3K27me3 on MHC-I promoter and joins CIITA as an NLR subfamily that controls MHC gene transcription.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Animals , B-Lymphocytes/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Histocompatibility Antigens Class I/genetics , Intracellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/metabolism , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Real-Time Polymerase Chain Reaction , T-Lymphocytes/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
12.
J Biol Chem ; 287(27): 23057-67, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22549783

ABSTRACT

Nucleotide binding and oligomerization domain-containing protein 2 (NOD2/Card15) is an intracellular protein that is involved in the recognition of bacterial cell wall-derived muramyl dipeptide. Mutations in the gene encoding NOD2 are associated with inherited inflammatory disorders, including Crohn disease and Blau syndrome. NOD2 is a member of the nucleotide-binding domain and leucine-rich repeat-containing protein gene (NLR) family. Nucleotide binding is thought to play a critical role in signaling by NLR family members. However, the molecular mechanisms underlying signal transduction by these proteins remain largely unknown. Mutations in the nucleotide-binding domain of NOD2 have been shown to alter its signal transduction properties in response to muramyl dipeptide in cellular assays. Using purified recombinant protein, we now demonstrate that NOD2 binds and hydrolyzes ATP. Additionally, we have found that the purified recombinant protein is able to bind directly to muramyl dipeptide and can associate with known NOD2-interacting proteins in vitro. Binding of NOD2 to muramyl dipeptide and homo-oligomerization of NOD2 are enhanced by ATP binding, suggesting a model of the molecular mechanism for signal transduction that involves binding of nucleotide followed by binding of muramyl dipeptide and oligomerization of NOD2 into a signaling complex. These findings set the stage for further studies into the molecular mechanisms that underlie detection of muramyl dipeptide and assembly of NOD2-containing signaling complexes.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Adenosine Triphosphate/metabolism , Immunity, Innate/physiology , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction/immunology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/analogs & derivatives , Animals , Bacterial Proteins/metabolism , Baculoviridae/genetics , Cells, Cultured , Chromatography, Affinity , HEK293 Cells , Humans , Insecta/cytology , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Protein Binding/physiology , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
13.
Immunity ; 36(5): 742-54, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22503542

ABSTRACT

In vitro data suggest that a subgroup of NLR proteins, including NLRP12, inhibits the transcription factor NF-κB, although physiologic and disease-relevant evidence is largely missing. Dysregulated NF-κB activity is associated with colonic inflammation and cancer, and we found Nlrp12(-/-) mice were highly susceptible to colitis and colitis-associated colon cancer. Polyps isolated from Nlrp12(-/-) mice showed elevated noncanonical NF-κB activation and increased expression of target genes that were associated with cancer, including Cxcl13 and Cxcl12. NLRP12 negatively regulated ERK and AKT signaling pathways in affected tumor tissues. Both hematopoietic- and nonhematopoietic-derived NLRP12 contributed to inflammation, but the latter dominantly contributed to tumorigenesis. The noncanonical NF-κB pathway was regulated upon degradation of TRAF3 and activation of NIK. NLRP12 interacted with both NIK and TRAF3, and Nlrp12(-/-) cells have constitutively elevated NIK, p100 processing to p52 and reduced TRAF3. Thus, NLRP12 is a checkpoint of noncanonical NF-κB, inflammation, and tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/pathology , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Intracellular Signaling Peptides and Proteins/metabolism , NF-kappa B/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Colitis/genetics , Colonic Neoplasms/genetics , Disease Progression , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , NF-kappaB-Inducing Kinase
14.
PLoS One ; 6(9): e24795, 2011.
Article in English | MEDLINE | ID: mdl-21966369

ABSTRACT

Plexins are cell surface receptors widely studied in the nervous system, where they mediate migration and morphogenesis though the Rho family of small GTPases. More recently, plexins have been implicated in immune processes including cell-cell interaction, immune activation, migration, and cytokine production. Plexin-B2 facilitates ligand induced cell guidance and migration in the nervous system, and induces cytoskeletal changes in overexpression assays through RhoGTPase. The function of Plexin-B2 in the immune system is unknown. This report shows that Plexin-B2 is highly expressed on cells of the innate immune system in the mouse, including macrophages, conventional dendritic cells, and plasmacytoid dendritic cells. However, Plexin-B2 does not appear to regulate the production of proinflammatory cytokines, phagocytosis of a variety of targets, or directional migration towards chemoattractants or extracellular matrix in mouse macrophages. Instead, Plxnb2(-/-) macrophages have greater cellular motility than wild type in the unstimulated state that is accompanied by more active, GTP-bound Rac and Cdc42. Additionally, Plxnb2(-/-) macrophages demonstrate faster in vitro wound closure activity. Studies have shown that a closely related family member, Plexin-B1, binds to active Rac and sequesters it from downstream signaling. The interaction of Plexin-B2 with Rac has only been previously confirmed in yeast and bacterial overexpression assays. The data presented here show that Plexin-B2 functions in mouse macrophages as a negative regulator of the GTPases Rac and Cdc42 and as a negative regulator of basal cell motility and wound healing.


Subject(s)
Cell Movement , Macrophages/metabolism , Nerve Tissue Proteins/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Blotting, Western , Bone Marrow Cells/metabolism , Cell Transplantation/methods , Cells, Cultured , Cytokines/metabolism , Female , Flow Cytometry , Humans , Leukocyte Common Antigens/metabolism , Liver/cytology , Liver/embryology , Liver/metabolism , Macrophages/cytology , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Phagocytosis , Protein Binding , Spleen/cytology , Spleen/metabolism
15.
Cell Host Microbe ; 10(1): 21-32, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21767809

ABSTRACT

The obligate intracellular bacterial pathogen Chlamydia trachomatis injects numerous effector proteins into the epithelial cell cytoplasm to manipulate host functions important for bacterial survival. In addition, the bacterium secretes a serine protease, chlamydial protease-like activity factor (CPAF). Although several CPAF targets are reported, the significance of CPAF-mediated proteolysis is unclear due to the lack of specific CPAF inhibitors and the diversity of host targets. We report that CPAF also targets chlamydial effectors secreted early during the establishment of the pathogen-containing vacuole ("inclusion"). We designed a cell-permeable CPAF-specific inhibitory peptide and used it to determine that CPAF prevents superinfection by degrading early Chlamydia effectors translocated during entry into a preinfected cell. Prolonged CPAF inhibition leads to loss of inclusion integrity and caspase-1-dependent death of infected epithelial cells. Thus, CPAF functions in niche protection, inclusion integrity and pathogen survival, making the development of CPAF-specific protease inhibitors an attractive antichlamydial therapeutic strategy.


Subject(s)
Chlamydia trachomatis/pathogenicity , Endopeptidases/metabolism , Host-Pathogen Interactions , Amino Acid Chloromethyl Ketones/pharmacology , Amino Acid Sequence , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Bacterial Secretion Systems , Caspase 1/metabolism , Cell Death/physiology , Cell Membrane Permeability , Chlamydia trachomatis/metabolism , Epithelial Cells/microbiology , Mice , Molecular Sequence Data , Peptides/metabolism , Protease Inhibitors/pharmacology , Protein Transport , Virulence Factors/metabolism
16.
Immunity ; 34(6): 854-65, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21703540

ABSTRACT

The nucleotide-binding domain and leucine-rich-repeat-containing (NLR) proteins regulate innate immunity. Although the positive regulatory impact of NLRs is clear, their inhibitory roles are not well defined. We showed that Nlrx1(-/-) mice exhibited increased expression of antiviral signaling molecules IFN-ß, STAT2, OAS1, and IL-6 after influenza virus infection. Consistent with increased inflammation, Nlrx1(-/-) mice exhibited marked morbidity and histopathology. Infection of these mice with an influenza strain that carries a mutated NS-1 protein, which normally prevents IFN induction by interaction with RNA and the intracellular RNA sensor RIG-I, further exacerbated IL-6 and type I IFN signaling. NLRX1 also weakened cytokine responses to the 2009 H1N1 pandemic influenza virus in human cells. Mechanistically, Nlrx1 deletion led to constitutive interaction of MAVS and RIG-I. Additionally, an inhibitory function is identified for NLRX1 during LPS activation of macrophages where the MAVS-RIG-I pathway was not involved. NLRX1 interacts with TRAF6 and inhibits NF-κB activation. Thus, NLRX1 functions as a checkpoint of overzealous inflammation.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Mitochondrial Proteins/immunology , Orthomyxoviridae Infections/immunology , Signal Transduction , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Interferon-beta/biosynthesis , Interferon-beta/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Macrophages/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , NF-kappa B/immunology , NF-kappa B/metabolism , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface , TNF Receptor-Associated Factor 6/immunology , TNF Receptor-Associated Factor 6/metabolism
17.
Blood ; 118(8): 2366-74, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21700772

ABSTRACT

Microparticles (MPs) are shed from activated and dying cells. They can transmit signals from cell to cell, locally or at a distance through the circulation. Monocytic MPs are elevated in different diseases, including bacterial infections. Here, we investigated how monocytic MPs activate endothelial cells. We found that MPs from lipopolysaccharide (LPS)-treated THP-1 monocytic cells bind to and are internalized by human endothelial cells. MPs from LPS-treated THP-1 cells, but not untreated cells, induced phosphorylation of ERK1/2, activation of the nuclear factor-κB pathway and expression of cell adhesion molecules intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin. Similar results were observed using MPs from LPS-treated peripheral blood mononuclear cells. We next investigated the mechanism by which monocytic MPs activated endothelial cells and found that they contain IL-1ß and components of the inflammasome, including apoptosis-associated speck-like protein containing a CARD, caspase-1, and NLRP3. Importantly, knockdown of NLRP3 in THP-1 cells reduced the activity of the MPs and blockade of the IL-1 receptor on endothelial cells decreased MP-dependent induction of cell adhesion molecules. Therefore, monocytic MPs contain IL-1ß and may amplify inflammation by enhancing the activation of the endothelium.


Subject(s)
Cell-Derived Microparticles/physiology , Endothelial Cells/physiology , Interleukin-1beta/physiology , Monocytes/physiology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Adhesion Molecules/physiology , Cell Line , Cell-Derived Microparticles/drug effects , Cells, Cultured , Gene Knockdown Techniques , Humans , Inflammasomes/physiology , Inflammation Mediators/physiology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Lipopolysaccharides/toxicity , Monocytes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1/antagonists & inhibitors , Signal Transduction
18.
Annu Rev Immunol ; 29: 707-35, 2011.
Article in English | MEDLINE | ID: mdl-21219188

ABSTRACT

Inflammasome activation leads to caspase-1 activation, which causes the maturation and secretion of pro-IL-1ß and pro-IL-18 among other substrates. A subgroup of the NLR (nucleotide-binding domain, leucine-rich repeat containing) proteins are key mediators of the inflammasome. Studies of gene-deficient mice and cells have implicated NLR inflammasomes in a host of responses to a wide range of microbial pathogens, inflammatory diseases, cancer, and metabolic and autoimmune disorders. Determining exactly how the inflammasome is activated in these diseases and disease models remains a challenge. This review presents and integrates recent progress in the field.


Subject(s)
Inflammasomes/immunology , Inflammation/immunology , Intercellular Signaling Peptides and Proteins/immunology , Animals , Humans , Inflammation Mediators/immunology , Metabolic Diseases/immunology , Neoplasms/immunology
19.
Science ; 331(6015): 330-4, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21252346

ABSTRACT

The NLR (nucleotide binding and oligomerization, leucine-rich repeat) family of proteins senses microbial infections and activates the inflammasome, a multiprotein complex that promotes microbial clearance. Kaposi's sarcoma-associated herpesvirus (KSHV) is linked to several human malignancies. We found that KSHV Orf63 is a viral homolog of human NLRP1. Orf63 blocked NLRP1-dependent innate immune responses, including caspase-1 activation and processing of interleukins IL-1ß and IL-18. KSHV Orf63 interacted with NLRP1, NLRP3, and NOD2. Inhibition of Orf63 expression resulted in increased expression of IL-1ß during the KSHV life cycle. Furthermore, inhibition of NLRP1 was necessary for efficient reactivation and generation of progeny virus. The viral homolog subverts the function of cellular NLRs, which suggests that modulation of NLR-mediated innate immunity is important for the lifelong persistence of herpesviruses.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Apoptosis Regulatory Proteins/antagonists & inhibitors , Herpesvirus 8, Human/physiology , Immune Evasion , Immunity, Innate , Inflammasomes/antagonists & inhibitors , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Apoptosis , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Caspase Inhibitors , Cell Line , Cell Line, Tumor , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Molecular Sequence Data , Monocytes/virology , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Proteins , Nod2 Signaling Adaptor Protein/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Transfection , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Activation , Virus Latency , Virus Replication
20.
J Immunol ; 186(3): 1333-7, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21191067

ABSTRACT

The nucleotide-binding domain leucine-rich repeat-containing proteins, NLRs, are intracellular sensors of pathogen-associated molecular patterns and damage-associated molecular patterns. A subgroup of NLRs can form inflammasome complexes, which facilitate the maturation of procaspase 1 to caspase 1, leading to IL-1ß and IL-18 cleavage and secretion. NLRC5 is predominantly expressed in hematopoietic cells and has not been studied for inflammasome function. RNA interference-mediated knockdown of NLRC5 nearly eliminated caspase 1, IL-1ß, and IL-18 processing in response to bacterial infection, pathogen-associated molecular patterns, and damage-associated molecular patterns. This was confirmed in primary human monocytic cells. NLRC5, together with procaspase 1, pro-IL-1ß, and the inflammasome adaptor ASC, reconstituted inflammasome activity that showed cooperativity with NLRP3. The range of pathogens that activate NLRC5 inflammasome overlaps with those that activate NLRP3. Furthermore, NLRC5 biochemically associates with NLRP3 in a nucleotide-binding domain-dependent but leucine-rich repeat-inhibitory fashion. These results invoke a model in which NLRC5 interacts with NLRP3 to cooperatively activate the inflammasome.


Subject(s)
Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Animals , Carrier Proteins/physiology , Cell Line, Transformed , Cell Line, Tumor , Escherichia coli/immunology , Escherichia coli/pathogenicity , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Inflammasomes/biosynthesis , Inflammasomes/genetics , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Leucine/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Structure, Tertiary/genetics , Shigella flexneri/immunology , Shigella flexneri/pathogenicity , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...