Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1155935, 2023.
Article in English | MEDLINE | ID: mdl-37325640

ABSTRACT

Introduction: Osteopontin (OPN; also known as SPP1), an immunomodulatory cytokine highly expressed in bone marrow-derived macrophages (BMMΦ), is known to regulate diverse cellular and molecular immune responses. We previously revealed that glatiramer acetate (GA) stimulation of BMMΦ upregulates OPN expression, promoting an anti-inflammatory, pro-healing phenotype, whereas OPN inhibition triggers a pro-inflammatory phenotype. However, the precise role of OPN in macrophage activation state is unknown. Methods: Here, we applied global proteome profiling via mass spectrometry (MS) analysis to gain a mechanistic understanding of OPN suppression versus induction in primary macrophage cultures. We analyzed protein networks and immune-related functional pathways in BMMΦ either with OPN knockout (OPNKO) or GA-mediated OPN induction compared with wild type (WT) macrophages. The most significant differentially expressed proteins (DEPs) were validated using immunocytochemistry, western blot, and immunoprecipitation assays. Results and discussion: We identified 631 DEPs in OPNKO or GA-stimulated macrophages as compared to WT macrophages. The two topmost downregulated DEPs in OPNKO macrophages were ubiquitin C-terminal hydrolase L1 (UCHL1), a crucial component of the ubiquitin-proteasome system (UPS), and the anti-inflammatory Heme oxygenase 1 (HMOX-1), whereas GA stimulation upregulated their expression. We found that UCHL1, previously described as a neuron-specific protein, is expressed by BMMΦ and its regulation in macrophages was OPN-dependent. Moreover, UCHL1 interacted with OPN in a protein complex. The effects of GA activation on inducing UCHL1 and anti-inflammatory macrophage profiles were mediated by OPN. Functional pathway analyses revealed two inversely regulated pathways in OPN-deficient macrophages: activated oxidative stress and lysosome-mitochondria-mediated apoptosis (e.g., ROS, Lamp1-2, ATP-synthase subunits, cathepsins, and cytochrome C and B subunits) and inhibited translation and proteolytic pathways (e.g., 60S and 40S ribosomal subunits and UPS proteins). In agreement with the proteome-bioinformatics data, western blot and immunocytochemical analyses revealed that OPN deficiency perturbs protein homeostasis in macrophages-inhibiting translation and protein turnover and inducing apoptosis-whereas OPN induction by GA restores cellular proteostasis. Taken together, OPN is essential for macrophage homeostatic balance via the regulation of protein synthesis, UCHL1-UPS axis, and mitochondria-mediated apoptotic processes, indicating its potential application in immune-based therapies.


Subject(s)
Osteopontin , Proteasome Endopeptidase Complex , Osteopontin/genetics , Osteopontin/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteostasis , Proteome/metabolism , Macrophages , Mitochondria/metabolism , Apoptosis
2.
J Transl Autoimmun ; 6: 100181, 2023.
Article in English | MEDLINE | ID: mdl-36619655

ABSTRACT

Rationale: Effective therapies to reduce the severity and high mortality of pulmonary vasculitis and diffuse alveolar hemorrhage (DAH) in patients with systemic lupus erythematosus (SLE) is a serious unmet need. We explored whether biologic neutralization of eNAMPT (extracellular nicotinamide phosphoribosyl-transferase), a novel DAMP and Toll-like receptor 4 ligand, represents a viable therapeutic strategy in lupus vasculitis. Methods: Serum was collected from SLE subjects (n = 37) for eNAMPT protein measurements. In the preclinical pristane-induced murine model of lung vasculitis/hemorrhage, C57BL/6 J mice (n = 5-10/group) were treated with PBS, IgG (1 mg/kg), or the eNAMPT-neutralizing ALT-100 mAb (1 mg/kg, IP or subcutaneously (SQ). Lung injury evaluation (Day 10) included histology/immuno-histochemistry, BAL protein/cellularity, tissue biochemistry, RNA sequencing, and plasma biomarker assessment. Results: SLE subjects showed highly significant increases in blood NAMPT mRNA expression and eNAMPT protein levels compared to healthy controls. Preclinical pristane-exposed mice studies showed significantly increased NAMPT lung tissue expression and increased plasma eNAMPT levels accompanied by marked increases in alveolar hemorrhage and lung inflammation (BAL protein, PMNs, activated monocytes). In contrast, ALT-100 mAb-treated mice showed significant attenuation of inflammatory lung injury, alveolar hemorrhage, BAL protein, tissue leukocytes, and plasma inflammatory cytokines (eNAMPT, IL-6, IL-8). Lung RNA sequencing showed pristane-induced activation of inflammatory genes/pathways including NFkB, cytokine/chemokine, IL-1ß, and MMP signaling pathways, each rectified in ALT-100 mAb-treated mice. Conclusions: These findings highlight the role of eNAMPT/TLR4-mediated inflammatory signaling in the pathobiology of SLE pulmonary vasculitis and alveolar hemorrhage. Biologic neutralization of this novel DAMP appears to serve as a viable strategy to reduce the severity of SLE lung vasculitis.

3.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33361152

ABSTRACT

The balance between NLRP3 inflammasome activation and mitophagy is essential for homeostasis and cellular health, but this relationship remains poorly understood. Here we found that interleukin-1α (IL-1α)-deficient macrophages have reduced caspase-1 activity and diminished IL-1ß release, concurrent with reduced mitochondrial damage, suggesting a role for IL-1α in regulating this balance. LPS priming of macrophages induced pro-IL-1α translocation to mitochondria, where it directly interacted with mitochondrial cardiolipin (CL). Computational modeling revealed a likely CL binding motif in pro-IL-1α, similar to that found in LC3b. Thus, binding of pro-IL-1α to CL in activated macrophages may interrupt CL-LC3b-dependent mitophagy, leading to enhanced Nlrp3 inflammasome activation and more robust IL-1ß production. Mutation of pro-IL-1α residues predicted to be involved in CL binding resulted in reduced pro-IL-1α-CL interaction, a reduction in NLRP3 inflammasome activity, and increased mitophagy. These data identify a function for pro-IL-1α in regulating mitophagy and the potency of NLRP3 inflammasome activation.


Subject(s)
Cardiolipins/metabolism , Interleukin-1alpha/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Autophagy , Cardiolipins/physiology , Caspase 1/metabolism , Female , HEK293 Cells , Humans , Inflammasomes/metabolism , Interleukin-1alpha/physiology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitophagy/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Protein Binding/physiology , Protein Domains/physiology , Reactive Oxygen Species/metabolism
4.
J Clin Invest ; 129(7): 2888-2897, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31038471

ABSTRACT

Influenza A virus (IAV)-specific T cell responses are important correlates of protection during primary and subsequent infections. Generation and maintenance of robust IAV-specific T cell responses relies on T cell interactions with dendritic cells (DCs). In this study, we explore the role of nucleotide-binding domain leucine-rich repeat containing receptor family member NLRC4 in modulating the DC phenotype during IAV infection. Nlrc4-/- mice had worsened survival and increased viral titers during infection, normal innate immune cell recruitment and IAV-specific CD8 T cell responses, but severely blunted IAV-specific CD4 T cell responses compared to wild-type mice. The defect in the pulmonary IAV-specific CD4 T cell response was not a result of defective priming or migration of these cells in Nlrc4-/- mice but was instead due to an increase in FasL+ DCs, resulting in IAV-specific CD4 T cell death. Together, our data support a novel role for NLRC4 in regulating the phenotype of lung DCs during a respiratory viral infection, and thereby influencing the magnitude of protective T cell responses.


Subject(s)
Apoptosis Regulatory Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/immunology , Dendritic Cells/immunology , Fas Ligand Protein/immunology , Gene Expression Regulation/immunology , Influenza A virus/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Animals , Apoptosis Regulatory Proteins/genetics , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/genetics , Dendritic Cells/pathology , Fas Ligand Protein/genetics , Lung/pathology , Mice , Mice, Knockout , Orthomyxoviridae Infections/pathology
5.
Front Immunol ; 10: 754, 2019.
Article in English | MEDLINE | ID: mdl-31031755

ABSTRACT

Autophagy can either antagonize or promote intracellular bacterial growth, depending on the pathogen. Here, we investigated the role of autophagy during a pulmonary infection with the obligate intracellular pathogen, Chlamydia pneumoniae (CP). In mouse embryonic fibroblasts (MEFs) or macrophages, deficiency of autophagy pathway components led to enhanced CP replication, suggesting that autophagy exerts a bactericidal role. However, in vivo, mice with myeloid-specific deletion of the autophagic protein ATG16L1 suffered increased mortality during CP infection, neutrophilia, and increased inflammasome activation despite no change in bacterial burden. Induction of autophagy led to reduced CP replication in vitro, but impaired survival in CP-infected mice, associated with an initial reduction in IL-1ß production, followed by enhanced neutrophil recruitment, defective CP clearance, and later inflammasome activation and IL-1ß production, which drove the resulting mortality. Taken together, our data suggest that a delicate interplay exists between autophagy and inflammasome activation in determining the outcome of CP infection, perturbation of which can result in inflammatory pathology or unrestricted bacterial growth.


Subject(s)
Autophagy , Chlamydophila Infections/metabolism , Chlamydophila Infections/microbiology , Chlamydophila pneumoniae/physiology , Inflammasomes/metabolism , Animals , Biomarkers , Fibroblasts/metabolism , Fibroblasts/microbiology , Flow Cytometry , Gene Knockout Techniques , Macrophages/metabolism , Macrophages/microbiology , Mice
6.
Cell Metab ; 28(3): 432-448.e4, 2018 09 04.
Article in English | MEDLINE | ID: mdl-29937375

ABSTRACT

Pathogen burden accelerates atherosclerosis, but the mechanisms remain unresolved. Activation of the NLRP3 inflammasome is linked to atherogenesis. Here we investigated whether Chlamydia pneumoniae (C.pn) infection engages NLRP3 in promoting atherosclerosis. C.pn potentiated hyperlipidemia-induced inflammasome activity in cultured macrophages and in foam cells in atherosclerotic lesions of Ldlr-/- mice. C.pn-induced acceleration of atherosclerosis was significantly dependent on NLRP3 and caspase-1. We discovered that C.pn-induced extracellular IL-1ß triggers a negative feedback loop to inhibit GPR109a and ABCA1 expression and cholesterol efflux, leading to accumulation of intracellular cholesterol and foam cell formation. Gpr109a and Abca1 were both upregulated in plaque lesions in Nlrp3-/- mice in both hyperlipidemic and C.pn infection models. Mature IL-1ß and cholesterol may compete for access to the ABCA1 transporter to be exported from macrophages. C.pn exploits this metabolic-immune crosstalk, which can be modulated by NLRP3 inhibitors to alleviate atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Atherosclerosis/microbiology , Chlamydophila pneumoniae/pathogenicity , Cholesterol/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Biological Transport , Caspase 1/metabolism , Female , Foam Cells/immunology , Foam Cells/pathology , Host Microbial Interactions , Inflammasomes , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/microbiology , Signal Transduction
7.
Arthritis Rheumatol ; 69(2): 410-421, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27696768

ABSTRACT

OBJECTIVE: Kawasaki disease (KD) is the leading cause of acquired heart disease among children in developed countries. Coronary lesions in KD in humans are characterized by an increased presence of infiltrating CD3+ T cells; however, the specific contributions of the different T cell subpopulations in coronary arteritis development remain unknown. Therefore, we sought to investigate the function of CD4+ and CD8+ T cells, Treg cells, and natural killer (NK) T cells in the pathogenesis of KD. METHODS: We addressed the function of T cell subsets in KD development by using a well-established murine model of Lactobacillus casei cell wall extract (LCWE)-induced KD vasculitis. We determined which T cell subsets were required for development of KD vasculitis by using several knockout murine strains and depleting monoclonal antibodies. RESULTS: LCWE-injected mice developed coronary lesions characterized by the presence of inflammatory cell infiltrates. Frequently, this chronic inflammation resulted in complete occlusion of the coronary arteries due to luminal myofibroblast proliferation (LMP) as well as the development of coronary arteritis and aortitis. We found that CD8+ T cells, but not CD4+ T cells, NK T cells, or Treg cells, were required for development of KD vasculitis. CONCLUSION: The LCWE-induced murine model of KD vasculitis mimics many histologic features of the disease in humans, such as the presence of CD8+ T cells and LMP in coronary artery lesions as well as epicardial coronary arteritis. Moreover, CD8+ T cells functionally contribute to the development of KD vasculitis in this murine model. Therapeutic strategies targeting infiltrating CD8+ T cells might be useful in the management of KD in humans.


Subject(s)
Arteritis/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Extracts/immunology , Cell Wall , Coronary Artery Disease/immunology , Lacticaseibacillus casei , Mucocutaneous Lymph Node Syndrome/immunology , Animals , Disease Models, Animal , Male , Mice, Inbred C57BL
8.
Circ Res ; 119(6): e76-90, 2016 Sep 02.
Article in English | MEDLINE | ID: mdl-27384322

ABSTRACT

RATIONALE: Activation of NLRP3 (nucleotide-binding domain and leucine-rich repeat pyrin domain containing 3) inflammasome-mediating interleukin (IL)-1ß secretion has emerged as an important component of inflammatory processes in atherosclerosis. Mitochondrial DNA (mtDNA) damage is detrimental in atherosclerosis, and mitochondria are central regulators of the nucleotide-binding domain and leucine-rich repeat pyrin domain containing 3 inflammasome. Human atherosclerotic plaques express increased mtDNA damage. The major DNA glycosylase, 8-oxoguanine glycosylase (OGG1), is responsible for removing the most abundant form of oxidative DNA damage. OBJECTIVE: To test the role of OGG1 in the development of atherosclerosis in mouse. METHODS AND RESULTS: We observed that Ogg1 expression decreases over time in atherosclerotic lesion macrophages of low-density lipoprotein receptor (Ldlr) knockout mice fed a Western diet. Ogg1(-/-)Ldlr(-/-) mice fed a Western diet resulted in an increase in plaque size and lipid content. We found increased oxidized mtDNA, inflammasome activation, and apoptosis in atherosclerotic lesions and also higher serum IL-1ß and IL-18 in Ogg1(-/-)Ldlr(-/-) mice than in Ldlr(-/-). Transplantation with Ogg1(-/-) bone marrow into Ldlr(-/-) mice led to larger atherosclerotic lesions and increased IL-1ß production. However, transplantation of Ogg1(-/-)Nlrp3(-/-) bone marrow reversed the Ogg1(-/-) phenotype of increased plaque size. Ogg1(-/-) macrophages showed increased oxidized mtDNA and had greater amounts of cytosolic mtDNA and cytochrome c, increased apoptosis, and more IL-1ß secretion. Finally, we found that proatherogenic miR-33 can directly inhibit human OGG1 expression and indirectly suppress both mouse and human OGG1 via AMP-activated protein kinase. CONCLUSIONS: OGG1 plays a protective role in atherogenesis by preventing excessive inflammasome activation. Our study provides insight into a new target for therapeutic intervention based on a link between oxidative mtDNA damage, OGG1, and atherosclerosis via NLRP3 inflammasome.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/prevention & control , DNA Glycosylases/metabolism , DNA Repair/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Atherosclerosis/genetics , DNA Glycosylases/deficiency , DNA Glycosylases/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Diet, Western/adverse effects , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
9.
Arterioscler Thromb Vasc Biol ; 35(12): 2605-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26515418

ABSTRACT

OBJECTIVE: Kawasaki disease (KD) is the most common cause of acute vasculitis and acquired cardiac disease among US children. We have previously shown that both TLR2/MyD88 and interleukin (IL)-1ß signaling are required for the Lactobacillus casei cell wall extract-induced KD vasculitis mouse model. The objectives of this study were to investigate the cellular origins of IL-1 production, the role of CD11c(+) dendritic cells and macrophages, and the relative contribution of hematopoietic and stromal cells for IL-1 responsive cells, as well the MyD88 signaling, in Lactobacillus casei cell wall extract-induced KD mouse model of vasculitis. APPROACH AND RESULTS: Using mouse knockout models and antibody depletion, we found that both IL-1α and IL-1ß were required for Lactobacillus casei cell wall extract-induced KD. Both dendritic cells and macrophages were necessary, and we found that MyD88 signaling was required in both hematopoietic and stromal cells. However, IL-1 response and signaling were critically required in nonendothelial stromal cells, but not in hematopoietic cells. CONCLUSIONS: Our results suggest that IL-1α and IL-1ß, as well as CD11c(+) dendritic cells and macrophages, are essential for the development of KD vasculitis and coronary arteritis in this mouse model. Bone marrow chimera experiments suggest that MyD88 signaling is important in both hematopoietic and stromal cells, whereas IL-1 signaling and response are required only in stromal cells, but not in endothelial cells. Determining the role of IL-1α and IL-1ß and of specific cell types in the KD vasculitis mouse model may have important implications for the design of more targeted therapies and understanding of the molecular mechanisms of KD immunopathologies.


Subject(s)
Aortitis/metabolism , Coronary Artery Disease/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Mucocutaneous Lymph Node Syndrome/metabolism , Signal Transduction , Stromal Cells/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortitis/chemically induced , Aortitis/genetics , Aortitis/pathology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Carrier Proteins/metabolism , Caspase 1/metabolism , Cell Wall , Cells, Cultured , Coronary Artery Disease/chemically induced , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Lacticaseibacillus casei , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mucocutaneous Lymph Node Syndrome/chemically induced , Mucocutaneous Lymph Node Syndrome/genetics , Mucocutaneous Lymph Node Syndrome/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Stromal Cells/pathology , Transplantation Chimera
10.
Innate Immun ; 21(7): 770-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26224488

ABSTRACT

The effect of TGF-ß1 on CpG DNA-induced type I IFN production was examined by reconstituting a series of signaling molecules in TLR 3 signaling. TGF-ß1 inhibited CpG DNA-induced IFN-α4 productivity in HeLa cells. Transfection of IFN regulatory factor (IRF)7 but not TNF receptor-associated factor (TRAF)6 and TRAF3 into cells triggered IFN-α4 productivity, and TGF-ß1 inhibited IRF7-mediated type I IFN production in the presence of TRAF6. TGF-ß1 induced ubiquitination of TRAF6, although CpG DNA did not induce it. Moreover, TGF-ß1 accelerated the ubiquitination of TRAF6 in the presence of CpG DNA. TGF-ß1 ubiquitinated TRAF6 at K63 but not K48. TGF-ß1 also induced ubiquitination of IRF7. Further, TGF-ß1 did not impair the interaction of IRF7 and TRAF6. CpG DNA induced the phosphorylation of IRF7 in the presence of TRAF6, whereas TGF-ß1 inhibited the IRF7 phosphorylation. Blocking of TRAF6 ubiquitination abolished the inhibition of CpG DNA-induced type I IFN production by TGF-ß. Taken together, TGF-ß was suggested to inhibit CpG DNA-induced type I IFN production transcriptionally via ubiquitination of TRAF6.


Subject(s)
Interferon-alpha/metabolism , Oligodeoxyribonucleotides/metabolism , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 6/metabolism , Transforming Growth Factor beta1/metabolism , DNA/genetics , HeLa Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon Regulatory Factor-7/metabolism , Phosphorylation , Protein Binding , Signal Transduction , Ubiquitination
11.
Immunity ; 42(4): 640-53, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25862090

ABSTRACT

Acute lung injury (ALI) remains a serious health issue with little improvement in our understanding of the pathophysiology and therapeutic approaches. We investigated the mechanism that lipopolysaccharide (LPS) induces early neutrophil recruitment to lungs and increases pulmonary vascular permeability during ALI. Intratracheal LPS induced release of pro-interleukin-1α (IL-1α) from necrotic alveolar macrophages (AM), which activated endothelial cells (EC) to induce vascular leakage via loss of vascular endothelial (VE)-cadherin. LPS triggered the AM purinergic receptor P2X7(R) to induce Ca(2+) influx and ATP depletion, which led to necrosis. P2X7R deficiency significantly reduced necrotic death of AM and release of pro-IL-1α into the lung. CD14 was required for LPS binding to P2X7R, as CD14 neutralization significantly diminished LPS induced necrotic death of AM and pro-IL-1α release. These results demonstrate a key role for pro-IL-1α from necrotic alveolar macrophages in LPS-mediated ALI, as a critical initiator of increased vascular permeability and early neutrophil infiltration.


Subject(s)
Interleukin-1alpha/immunology , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/drug effects , Receptors, Purinergic P2X7/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Adenosine Triphosphate/metabolism , Animals , Cadherins/genetics , Cadherins/immunology , Calcium/metabolism , Capillary Permeability/immunology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/pathology , Gene Expression Regulation , HEK293 Cells , Humans , Interleukin-1alpha/genetics , Intubation, Intratracheal , Lipopolysaccharide Receptors/genetics , Lung/drug effects , Lung/immunology , Lung/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Mice , Mice, Transgenic , Necrosis/chemically induced , Necrosis/immunology , Necrosis/pathology , Neutrophil Infiltration , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Protein Precursors/genetics , Protein Precursors/immunology , Receptors, Purinergic P2X7/genetics , Signal Transduction
12.
J Immunol ; 194(4): 1686-94, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25576596

ABSTRACT

We previously identified a novel alternatively spliced isoform of human myeloid differentiation protein-2 (MD-2s) that competitively inhibits binding of MD-2 to TLR4 in vitro. In this study, we investigated the protective role of MD-2s in LPS-induced acute lung injury by delivering intratracheally an adenovirus construct that expressed MD-2s (Ad-MD-2s). After adenovirus-mediated gene transfer, MD-2s was strongly expressed in lung epithelial cells and readily detected in bronchoalveolar lavage fluid. Compared to adenovirus serotype 5 containing an empty vector lacking a transgene control mice, Ad-MD-2s delivery resulted in significantly less LPS-induced inflammation in the lungs, including less protein leakage, cell recruitment, and expression of proinflammatory cytokines and chemokines, such as IL-6, keratinocyte chemoattractant, and MIP-2. Bronchoalveolar lavage fluid from Ad-MD-2s mice transferred into lungs of naive mice before intratracheal LPS challenge diminished proinflammatory cytokine levels. As house dust mite (HDM) sensitization is dependent on TLR4 and HDM Der p 2, a structural homolog of MD-2, we also investigated the effect of MD-2s on HDM-induced allergic airway inflammation. Ad-MD-2s given before HDM sensitization significantly inhibited subsequent allergic airway inflammation after HDM challenge, including reductions in eosinophils, goblet cell hyperplasia, and IL-5 levels. Our study indicates that the alternatively spliced short isoform of human MD-2 could be a potential therapeutic candidate to treat human diseases induced or exacerbated by TLR4 signaling, such as Gram-negative bacterial endotoxin-induced lung injury and HDM-triggered allergic lung inflammation.


Subject(s)
Lymphocyte Antigen 96/immunology , Pneumonia/genetics , Pneumonia/immunology , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Alternative Splicing , Animals , Blotting, Western , Disease Models, Animal , Female , Flow Cytometry , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Immunohistochemistry , Lymphocyte Antigen 96/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Isoforms/genetics , Protein Isoforms/immunology , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 4/immunology , Transfection
13.
Immunopharmacol Immunotoxicol ; 36(3): 237-41, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24852317

ABSTRACT

The effect of spironolactone (SPIR) on lipopolysaccharide (LPS)-induced production of proinflammatory mediators was examined using RAW 264.7 macrophage-like cells and mouse peritoneal macrophages. SPIR significantly inhibited LPS-induced production of nitric oxide (NO), tumor necrosis factor-α and prostaglandin E2. The inhibition was not mediated by cell death. SPIR reduced the expression of an inducible NO synthase mRNA in response to LPS. SPIR significantly inhibited phosphorylation of p65 nuclear factor (NF)-κB in response to LPS. Furthermore, SPIR inhibited phosphorylation of IκB kinase (IKK) as an upstream molecule of NF-κB in response to LPS. LPS did not induce the production of aldosterone in RAW 264.7 cells. Taken together, SPIR is suggested to inhibit LPS-induced proinflammatory mediators via inactivation of IKK/NF-κB in LPS signaling.


Subject(s)
Inflammation Mediators/antagonists & inhibitors , Lipopolysaccharides/pharmacology , NF-kappa B/antagonists & inhibitors , Spironolactone/pharmacology , Aldosterone/biosynthesis , Animals , Cells, Cultured , Dinoprostone/biosynthesis , I-kappa B Kinase/metabolism , Inflammation Mediators/metabolism , Mice , Nitric Oxide/biosynthesis , Phosphorylation , Tumor Necrosis Factor-alpha/biosynthesis
14.
Am J Respir Cell Mol Biol ; 50(2): 270-80, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24007300

ABSTRACT

IL-1ß is a potent proinflammatory cytokine that is implicated in the pathogenesis of acute respiratory distress syndrome. We hypothesized that LPS and mechanical ventilation (MV) together could lead to IL-1ß secretion and the development of acute lung injury (ALI), and that this process would be dependent on caspase-1 and the nucleotide binding domain and leucine-rich repeat (NLR) pyrin domain containing 3 (NLRP3) inflammasome activation. The objectives of this study were to determine the specific role of IL-1ß, caspase-1, and the NLRP3 inflammasome in a two-hit model of ALI due to LPS plus MV. We used a two-hit murine model of ALI in which both inhaled LPS and MV were required for the development of hypoxemia, pulmonary neutrophil infiltration, and alveolar leakage. Nlrp3-deficent and Casp1-deficient mice had significantly diminished IL-1ß levels in bronchoalveolar lavage fluid, and were specifically protected from hypoxemia, despite similar alveolar neutrophil infiltration and leakage. The IL-1 receptor antagonist, Anakinra, significantly improved the specific development of hypoxemia without significant effects on neutrophil infiltration or alveolar leakage. MV resulted in increased bronchoalveolar lavage extracellular ATP and alveolar macrophage apoptosis as triggers of NLRP3 inflammasome activation. NLRP3 inflammasome activation and IL-1ß production play a key role in ALI caused by the combination of LPS and MV, particularly in the hypoxemia associated with acute respiratory distress syndrome. Blocking IL-1 signaling in this model specifically ameliorates hypoxemia, without affecting neutrophil infiltration and alveolar leakage, disassociating these readouts of ALI. MV causes alveolar macrophage apoptosis, a key step in the activation of NLRP3 inflammasome and production of IL-1ß.


Subject(s)
Acute Lung Injury/metabolism , Carrier Proteins/metabolism , Hypoxia/immunology , Neutrophil Infiltration/immunology , Acute Lung Injury/immunology , Animals , Caspase 1/immunology , Caspase 1/metabolism , Disease Models, Animal , Inflammation/immunology , Interleukin-1/immunology , Interleukin-1/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Respiration, Artificial/adverse effects , Respiration, Artificial/methods , Signal Transduction/immunology
15.
Immunity ; 36(3): 401-14, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22342844

ABSTRACT

We report that in the presence of signal 1 (NF-κB), the NLRP3 inflammasome was activated by mitochondrial apoptotic signaling that licensed production of interleukin-1ß (IL-1ß). NLRP3 secondary signal activators such as ATP induced mitochondrial dysfunction and apoptosis, resulting in release of oxidized mitochondrial DNA (mtDNA) into the cytosol, where it bound to and activated the NLRP3 inflammasome. The antiapoptotic protein Bcl-2 inversely regulated mitochondrial dysfunction and NLRP3 inflammasome activation. Mitochondrial DNA directly induced NLRP3 inflammasome activation, because macrophages lacking mtDNA had severely attenuated IL-1ß production, yet still underwent apoptosis. Both binding of oxidized mtDNA to the NLRP3 inflammasome and IL-1ß secretion could be competitively inhibited by the oxidized nucleoside 8-OH-dG. Thus, our data reveal that oxidized mtDNA released during programmed cell death causes activation of the NLRP3 inflammasome. These results provide a missing link between apoptosis and inflammasome activation, via binding of cytosolic oxidized mtDNA to the NLRP3 inflammasome.


Subject(s)
Apoptosis/immunology , Carrier Proteins/immunology , Carrier Proteins/metabolism , DNA, Mitochondrial/immunology , DNA, Mitochondrial/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Animals , Gene Expression , Interleukin-1beta/biosynthesis , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidation-Reduction , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Signal Transduction
16.
J Immunol ; 187(8): 4331-7, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21918188

ABSTRACT

Stimulation with LPS induces tyrosine phosphorylation of numerous proteins involved in the TLR signaling pathway. In this study, we demonstrated that myeloid differentiation factor-2 (MD-2) is also tyrosine phosphorylated following LPS stimulation. LPS-induced tyrosine phosphorylation of MD-2 is specific; it is blocked by the tyrosine kinase inhibitor, herbimycin A, as well as by an inhibitor of endocytosis, cytochalasin D, suggesting that MD-2 phosphorylation occurs during trafficking of MD-2 and not on the cell surface. Furthermore, we identified two possible phospho-accepting tyrosine residues at positions 22 and 131. Mutant proteins in which these tyrosines were changed to phenylalanine had reduced phosphorylation and significantly diminished ability to activate NF-κB in response to LPS. In addition, MD-2 coprecipitated and colocalized with Lyn kinase, most likely in the endoplasmic reticulum. A Lyn-binding peptide inhibitor abolished MD-2 tyrosine phosphorylation, suggesting that Lyn is a likely candidate to be the kinase required for MD-2 tyrosine phosphorylation. Our study demonstrated that tyrosine phosphorylation of MD-2 is important for signaling following exposure to LPS and underscores the importance of this event in mediating an efficient and prompt immune response.


Subject(s)
Lymphocyte Antigen 96/metabolism , Phosphotyrosine/metabolism , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , src-Family Kinases/metabolism , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Lipopolysaccharides/immunology , Lymphocyte Antigen 96/immunology , Microscopy, Fluorescence , Toll-Like Receptor 4/metabolism , src-Family Kinases/immunology
17.
Innate Immun ; 17(1): 97-105, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20023007

ABSTRACT

ONO 3403, a new synthetic serine protease inhibitor, is a derivative of camostat mesilate and has a higher protease-inhibitory activity. The effect of ONO 3403 on lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF)-α and nitric oxide (NO) production in RAW 264.7 macrophage-like cells was examined. ONO 3403 significantly inhibited LPS-induced TNF-α production at a lower concentration than camostat mesilate. It also inhibited LPS-induced NO production. Their inhibition was responsible for the reduced mRNA expression of TNF-α and inducible NO synthase. In LPS-stimulated cells, ONO 3403 prevented the augmentation of MyD88 expression and inhibited the phosphorylation of IκB-α, stress-activated protein kinase (SAPK) and IRF-3, and the production of interferon-ß. ONO 3403 abolished the elevation of the extracellular serine protease activity in response to LPS. Further, it reduced the circulating TNF-α level, hepatic injury and mortality in mice receiving an injection of D-galactosamine and LPS. ONO 3403 was suggested to inhibit LPS-induced inflammatory responses via inactivation of MyD88-dependent and independent pathways.


Subject(s)
Allylglycine/analogs & derivatives , Benzamidines/pharmacology , Benzamidines/therapeutic use , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Nitric Oxide/metabolism , Shock, Septic/prevention & control , Tumor Necrosis Factor-alpha/metabolism , Allylglycine/pharmacology , Allylglycine/therapeutic use , Animals , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Esters , Female , Gabexate/analogs & derivatives , Gabexate/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Guanidines , I-kappa B Proteins/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-gamma/blood , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Liver/pathology , Macrophages/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/metabolism , NF-KappaB Inhibitor alpha , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Serine Proteases/metabolism , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use , Shock, Septic/blood , Shock, Septic/pathology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
18.
Cancer Invest ; 28(8): 806-12, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20594067

ABSTRACT

The role of retinoblastoma protein-interacting zinc finger 1 (RIZ1) on the cell growth of mouse and human monocytic leukemia cells was examined. RIZ1 expression was induced in response to tumor necrosis factor (TNF)-α. The expression was dependent on the nuclear factor-κB and AKT signaling. Further, RIZ1 expression led to the augmentation of p53 expression and the silencing of RIZ1 prevented it. On the other hand, a p53 inhibitor enhanced the TNF-α-induced RIZ1 expression. Silencing of RIZ1 augmented the proliferative activity of TNF-α-treated cells. Therefore, it is suggested that RIZ1 negatively regulated the cell proliferation of monocytic leukemia cells via activation of p53.


Subject(s)
DNA-Binding Proteins/pharmacology , Histone-Lysine N-Methyltransferase/pharmacology , Leukemia, Myeloid/pathology , Nuclear Proteins/pharmacology , Transcription Factors/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Division/drug effects , DNA Methylation , DNA Primers , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone-Lysine N-Methyltransferase/genetics , Homeostasis , Humans , Leukemia, Myeloid/genetics , Mice , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Suppressor Protein p53/drug effects
19.
Cell Immunol ; 264(2): 114-8, 2010.
Article in English | MEDLINE | ID: mdl-20557878

ABSTRACT

The involvement of retinoblastoma protein-interacting zinc finger 1 (RIZ1), a tumor suppressor, in lipopolysaccharide (LPS)-induced inflammatory responses was investigated by using RAW 264.7 macrophage-like cells. LPS significantly augmented the expression of RIZ1 and the augmentation was mediated by the activation of nuclear factor (NF)-kappaB and Akt. The silencing of RIZ1 with the siRNA led to the inactivation of NF-kappaB in response to LPS. Moreover, the RIZ1 silencing caused the down-regulation of p53 activation and a p53 pharmacological inhibitor attenuated the RIZ1 expression. LPS-induced tumor necrosis factor-alpha and interleukin-6 production was prevented by RIZ1 siRNA or a p53 pharmacological inhibitor. Therefore, RIZ1 was suggested to augment LPS-induced NF-kappaB activation in collaboration with p53 and enhance the production of proinflammatory cytokines in response to LPS.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Interleukin-6/biosynthesis , Macrophages/metabolism , NF-kappa B/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Line , Cloning, Molecular , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/immunology , Inflammation , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Macrophages/immunology , Macrophages/pathology , Mice , Mutation/genetics , NF-kappa B/genetics , RNA, Small Interfering/genetics , Transcription Factors/genetics , Transcription Factors/immunology , Transcriptional Activation/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Immunology ; 131(1): 59-66, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20406300

ABSTRACT

Selective Alzheimer disease indicator-1 (seladin-1) is a broadly expressed oxidoreductase and is related to Alzheimer disease, cholesterol metabolism and carcinogenesis. The effect of lipopolysaccharide (LPS) on the expression of seladin-1 was examined using RAW 264.7 macrophage-like cells and murine peritoneal macrophages. Lipopolysaccharide induced the expression of seladin-1 protein and messenger RNA in those macrophages. The seladin-1 expression was also augmented by a series of Toll-like receptor ligands. The LPS augmented the expression of seladin-1 via reactive oxygen species generation and p38 activation. Seladin-1 inhibited LPS-induced activation of p38 but not nuclear factor-kappaB and inhibited the production of tumour necrosis factor-alpha in response to LPS. Moreover, seladin-1 inhibited LPS-induced osteoclast formation and enhanced LPS-induced alkaline phosphatase activity. Therefore, it was suggested that seladin-1 might be an LPS-responsible gene product and regulate the LPS-induced inflammatory response negatively.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/drug effects , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Cell Line , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Osteoclasts/cytology , Osteoclasts/metabolism , Oxidoreductases Acting on CH-CH Group Donors/drug effects , Oxidoreductases Acting on CH-CH Group Donors/genetics , Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...