Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Immunity ; 54(7): 1463-1477.e11, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34115964

ABSTRACT

Acute respiratory distress syndrome (ARDS), an inflammatory condition with high mortality rates, is common in severe COVID-19, whose risk is reduced by metformin rather than other anti-diabetic medications. Detecting of inflammasome assembly in post-mortem COVID-19 lungs, we asked whether and how metformin inhibits inflammasome activation while exerting its anti-inflammatory effect. We show that metformin inhibited NLRP3 inflammasome activation and interleukin (IL)-1ß production in cultured and alveolar macrophages along with inflammasome-independent IL-6 secretion, thus attenuating lipopolysaccharide (LPS)- and SARS-CoV-2-induced ARDS. By targeting electron transport chain complex 1 and independently of AMP-activated protein kinase (AMPK) or NF-κB, metformin blocked LPS-induced and ATP-dependent mitochondrial (mt) DNA synthesis and generation of oxidized mtDNA, an NLRP3 ligand. Myeloid-specific ablation of LPS-induced cytidine monophosphate kinase 2 (CMPK2), which is rate limiting for mtDNA synthesis, reduced ARDS severity without a direct effect on IL-6. Thus, inhibition of ATP and mtDNA synthesis is sufficient for ARDS amelioration.


Subject(s)
Adenosine Triphosphate/metabolism , DNA, Mitochondrial/biosynthesis , Inflammasomes/drug effects , Metformin/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pneumonia/prevention & control , Animals , COVID-19/metabolism , COVID-19/prevention & control , Cytokines/genetics , Cytokines/metabolism , DNA, Mitochondrial/metabolism , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Metformin/therapeutic use , Mice , Nucleoside-Phosphate Kinase/metabolism , Pneumonia/metabolism , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/prevention & control , SARS-CoV-2/pathogenicity
2.
Immunity ; 51(3): 508-521.e6, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31471109

ABSTRACT

Recent experimental data and clinical, genetic, and transcriptome evidence from patients converge to suggest a key role of interleukin-1ß (IL-1ß) in the pathogenesis of Kawasaki disease (KD). However, the molecular mechanisms involved in the development of cardiovascular lesions during KD vasculitis are still unknown. Here, we investigated intestinal barrier function in KD vasculitis and observed evidence of intestinal permeability and elevated circulating secretory immunoglobulin A (sIgA) in KD patients, as well as elevated sIgA and IgA deposition in vascular tissues in a mouse model of KD vasculitis. Targeting intestinal permeability corrected gut permeability, prevented IgA deposition and ameliorated cardiovascular pathology in the mouse model. Using genetic and pharmacologic inhibition of IL-1ß signaling, we demonstrate that IL-1ß lies upstream of disrupted intestinal barrier function, subsequent IgA vasculitis development, and cardiac inflammation. Targeting mucosal barrier dysfunction and the IL-1ß pathway may also be applicable to other IgA-related diseases, including IgA vasculitis and IgA nephropathy.


Subject(s)
Cardiovascular Diseases/immunology , Immunoglobulin A/immunology , Inflammation/immunology , Intestines/immunology , Animals , Disease Models, Animal , Humans , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Mucocutaneous Lymph Node Syndrome/immunology , Permeability , Signal Transduction/immunology , Vasculitis/immunology
3.
Immunity ; 49(5): 873-885.e7, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30366765

ABSTRACT

Receptor interacting protein 2 (RIP2) plays a role in sensing intracellular pathogens, but its function in T cells is unclear. We show that RIP2 deficiency in CD4+ T cells resulted in chronic and severe interleukin-17A-mediated inflammation during Chlamydia pneumoniae lung infection, increased T helper 17 (Th17) cell formation in lungs of infected mice, accelerated atherosclerosis, and more severe experimental autoimmune encephalomyelitis. While RIP2 deficiency resulted in reduced conventional Th17 cell differentiation, it led to significantly enhanced differentiation of pathogenic (p)Th17 cells, which was dependent on RORα transcription factor and interleukin-1 but independent of nucleotide oligomerization domain (NOD) 1 and 2. Overexpression of RIP2 resulted in suppression of pTh17 cell differentiation, an effect mediated by its CARD domain, and phenocopied by a cell-permeable RIP2 CARD peptide. Our data suggest that RIP2 has a T cell-intrinsic role in determining the balance between homeostatic and pathogenic Th17 cell responses.


Subject(s)
Cell Differentiation/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Atherosclerosis , Biomarkers , Caspase Activation and Recruitment Domain , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/mortality , Gene Expression , Immunophenotyping , Inflammation/genetics , Inflammation/metabolism , Interleukin-17/biosynthesis , Interleukin-1beta , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
4.
Immunity ; 43(2): 213-5, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26287676

ABSTRACT

Inhibition of the inflammasome might be beneficial for numerous inflammatory pathologies. In this issue of Immunity, de Almeida et al. (2015) report that the PYRIN domain-only protein (POP1) efficiently inhibits inflammasome activation, identifying it as a pan-inflammasome inhibitor.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/immunology , Dendritic Cells/immunology , Inflammasomes/metabolism , Macrophages, Peritoneal/immunology , Monocytes/immunology , Peritonitis/immunology , Ribonucleoproteins/metabolism , Animals , Female , Humans
5.
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
6.
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
8.
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
9.
PLoS Pathog ; 14(9): e1007260, 2018 09.
Article in English | MEDLINE | ID: mdl-30235351

ABSTRACT

The gastrointestinal microbiota influences immune function throughout the body. The gut-lung axis refers to the concept that alterations of gut commensal microorganisms can have a distant effect on immune function in the lung. Overgrowth of intestinal Candida albicans has been previously observed to exacerbate allergic airways disease in mice, but whether subtler changes in intestinal fungal microbiota can affect allergic airways disease is less clear. In this study we have investigated the effects of the population expansion of commensal fungus Wallemia mellicola without overgrowth of the total fungal community. Wallemia spp. are commonly found as a minor component of the commensal gastrointestinal mycobiota in both humans and mice. Mice with an unaltered gut microbiota community resist population expansion when gavaged with W. mellicola; however, transient antibiotic depletion of gut microbiota creates a window of opportunity for expansion of W. mellicola following delivery of live spores to the gastrointestinal tract. This phenomenon is not universal as other commensal fungi (Aspergillus amstelodami, Epicoccum nigrum) do not expand when delivered to mice with antibiotic-depleted microbiota. Mice with Wallemia-expanded gut mycobiota experienced altered pulmonary immune responses to inhaled aeroallergens. Specifically, after induction of allergic airways disease with intratracheal house dust mite (HDM) antigen, mice demonstrated enhanced eosinophilic airway infiltration, airway hyperresponsiveness (AHR) to methacholine challenge, goblet cell hyperplasia, elevated bronchoalveolar lavage IL-5, and enhanced serum HDM IgG1. This phenomenon occurred with no detectable Wallemia in the lung. Targeted amplicon sequencing analysis of the gastrointestinal mycobiota revealed that expansion of W. mellicola in the gut was associated with additional alterations of bacterial and fungal commensal communities. We therefore colonized fungus-free Altered Schaedler Flora (ASF) mice with W. mellicola. ASF mice colonized with W. mellicola experienced enhanced severity of allergic airways disease compared to fungus-free control ASF mice without changes in bacterial community composition.


Subject(s)
Basidiomycota/immunology , Basidiomycota/pathogenicity , Gastrointestinal Microbiome/immunology , Mycobiome/immunology , Respiratory Hypersensitivity/etiology , Allergens/administration & dosage , Animals , Anti-Bacterial Agents/adverse effects , Antigens, Dermatophagoides/administration & dosage , Basidiomycota/growth & development , Disease Models, Animal , Environmental Microbiology , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Germ-Free Life/immunology , Humans , Mice , Mice, Inbred C57BL , Mycobiome/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/microbiology , Symbiosis/immunology
10.
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
11.
Arterioscler Thromb Vasc Biol ; 36(5): 886-97, 2016 05.
Article in English | MEDLINE | ID: mdl-26941015

ABSTRACT

OBJECTIVE: Kawasaki disease (KD) is the most common cause of acquired cardiac disease in US children. In addition to coronary artery abnormalities and aneurysms, it can be associated with systemic arterial aneurysms. We evaluated the development of systemic arterial dilatation and aneurysms, including abdominal aortic aneurysm (AAA) in the Lactobacillus casei cell-wall extract (LCWE)-induced KD vasculitis mouse model. METHODS AND RESULTS: We discovered that in addition to aortitis, coronary arteritis and myocarditis, the LCWE-induced KD mouse model is also associated with abdominal aorta dilatation and AAA, as well as renal and iliac artery aneurysms. AAA induced in KD mice was exclusively infrarenal, both fusiform and saccular, with intimal proliferation, myofibroblastic proliferation, break in the elastin layer, vascular smooth muscle cell loss, and inflammatory cell accumulation in the media and adventitia. Il1r(-/-), Il1a(-/-), and Il1b(-/-) mice were protected from KD associated AAA. Infiltrating CD11c(+) macrophages produced active caspase-1, and caspase-1 or NLRP3 deficiency inhibited AAA formation. Treatment with interleukin (IL)-1R antagonist (Anakinra), anti-IL-1α, or anti-IL-1ß mAb blocked LCWE-induced AAA formation. CONCLUSIONS: Similar to clinical KD, the LCWE-induced KD vasculitis mouse model can also be accompanied by AAA formation. Both IL-1α and IL-1ß play a key role, and use of an IL-1R blocking agent that inhibits both pathways may be a promising therapeutic target not only for KD coronary arteritis, but also for the other systemic arterial aneurysms including AAA that maybe seen in severe cases of KD. The LCWE-induced vasculitis model may also represent an alternative model for AAA disease.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Mucocutaneous Lymph Node Syndrome/complications , Receptors, Interleukin-1 Type I/metabolism , Signal Transduction , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , Aortitis/genetics , Aortitis/metabolism , Aortitis/pathology , Caspase 1/deficiency , Caspase 1/genetics , Cell Proliferation , Cell Wall , Dilatation, Pathologic , Disease Models, Animal , Elastin/metabolism , Female , Gene Expression Profiling , Genotype , Humans , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1alpha/deficiency , Interleukin-1alpha/genetics , Interleukin-1beta/deficiency , Interleukin-1beta/genetics , Lacticaseibacillus casei , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mucocutaneous Lymph Node Syndrome/chemically induced , Mucocutaneous Lymph Node Syndrome/drug therapy , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phenotype , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Signal Transduction/drug effects , Time Factors
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.
J Immunol ; 194(8): 3840-51, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25754739

ABSTRACT

Mast cells are known as central players in allergy and anaphylaxis, and they play a pivotal role in host defense against certain pathogens. Chlamydia pneumoniae is an important human pathogen, but it is unclear what role mast cells play during C. pneumoniae infection. We infected C57BL/6 (wild-type [WT]) and mast cell-deficient mice (Kit(W-sh/W-sh) [Wsh]) with C. pneumoniae. Wsh mice showed improved survival compared with WT mice, with fewer cells in Wsh bronchoalveolar lavage fluid (BALF), despite similar levels of cytokines and chemokines. We also found a more rapid clearance of bacteria from the lungs of Wsh mice compared with WT mice. Cromolyn, a mast cell stabilizer, reduced BALF cells and bacterial burden similar to the levels seen in Wsh mice; conversely, Compound 48/80, a mast cell degranulator, increased the number of BALF cells and bacterial burden. Histology showed that WT lungs had diffuse inflammation, whereas Wsh mice had patchy accumulations of neutrophils and perivascular accumulations of lymphocytes. Infected Wsh mice had reduced amounts of matrix metalloprotease-9 in BALF and were resistant to epithelial integral membrane protein degradation, suggesting that barrier integrity remains intact in Wsh mice. Mast cell reconstitution in Wsh mice led to enhanced bacterial growth and normal epithelial integral membrane protein degradation, highlighting the specific role of mast cells in this model. These data suggest that mast cells play a detrimental role during C. pneumoniae infection by facilitating immune cell infiltration into the airspace and providing a more favorable replicative environment for C. pneumoniae.


Subject(s)
Cell Movement/immunology , Chlamydophila Infections/immunology , Chlamydophila pneumoniae/immunology , Mast Cells/immunology , Pneumonia, Bacterial/immunology , Animals , Anti-Asthmatic Agents/pharmacology , Bronchoalveolar Lavage Fluid , Cell Movement/drug effects , Cell Movement/genetics , Chlamydophila Infections/genetics , Chlamydophila Infections/pathology , Cromolyn Sodium/pharmacology , Humans , Mast Cells/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/immunology , Mice , Mice, Transgenic , Pneumonia, Bacterial/genetics , Proteolysis/drug effects , p-Methoxy-N-methylphenethylamine/pharmacology
14.
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
15.
Curr Opin Pediatr ; 28(6): 764-771, 2016 12.
Article in English | MEDLINE | ID: mdl-27606957

ABSTRACT

PURPOSE OF REVIEW: Asthma is a complex and heterogeneous disease with strong genetic and environmental components that manifests within a variety of clinical features and diverse patterns of immune responses. Asthma prevalence has dramatically increased over the last decade in Westernized societies, thereby suggesting a key function of environmental factors in disease promotion and development. RECENT FINDINGS: 'Early-life' microbial exposure and bacterial colonization are crucial for the maturation and the education of the immune system. The commensal flora is also critical in order to maintain immune homeostasis at the mucosal surfaces and may consequently play an important function in allergic disease development. Recent evidence demonstrates that asthma influences and is also impacted by the composition and function of the human intestinal and respiratory microbiome. SUMMARY: In this review, we summarize the most recent findings on how asthma development is connected with respiratory and intestinal microbial dysbiosis. We highlight and discuss recent research that reveals the existence of a 'gut-lung' microbial axis and its impact on asthma development. We also analyze how 'early-life' microbial exposure affects the immune response and the consequences for asthma development.


Subject(s)
Asthma/microbiology , Dysbiosis/complications , Intestines/microbiology , Microbiota/immunology , Respiratory System/microbiology , Asthma/immunology , Dysbiosis/immunology , Humans , Intestines/immunology , Respiratory System/immunology , Risk Factors
16.
Cell Immunol ; 294(1): 25-32, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25666507

ABSTRACT

Several studies have demonstrated a strong link between Chlamydia pneumoniae (Cp) infection and atherosclerosis progression/exacerbation. Here, we try to understand whether a single administration of Cp could exacerbate atherosclerosis. Apoe(-/-) mice were intranasally infected with Cp followed by a high fat diet. Mice were sacrificed at different time points after Cp infection to monitor the development of the atheroma. Cp infection increased lipid content in the aortic sinus of Apoe(-/-) mice starting from 8 weeks. This was associated with increased numbers of active myeloid dendritic cells and plasmacytoid DCs which were co-localized with T-cells in the atherosclerotic plaque. The serum levels of IFN-γ showed a Th1-like environment typical of atherosclerosis. In conclusion, we demonstrate that one dose of Cp could exacerbate atherosclerotic lesion development, triggering innate immune cell accumulation early on that allowed the involvement of Th1-like cells in the exacerbation of the atherosclerotic plaque at later time points.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/immunology , Chlamydophila Infections/immunology , Plaque, Atherosclerotic/immunology , Th1 Cells/immunology , Animals , Chlamydophila Infections/microbiology , Chlamydophila pneumoniae/immunology , Dendritic Cells/immunology , Disease Progression , Interferon-gamma/blood , Lipids/biosynthesis , Mice , Mice, Inbred C57BL , Plaque, Atherosclerotic/microbiology , Sinus of Valsalva/microbiology , Sinus of Valsalva/pathology
17.
J Immunol ; 190(5): 2391-402, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23355734

ABSTRACT

The antitumor activity of LPS was first described by Dr. William Coley. However, its role in lung cancer remains unclear. The aim of our study was to elucidate the dose-dependent effects of LPS (0.1-10 µg/mouse) in a mouse model of B16-F10-induced metastatic lung cancer. Lung tumor growth increased at 3 and 7 d after the administration of low-dose LPS (0.1 µg/mouse) compared with control mice. This was associated with an influx of plasmacytoid dendritic cells (pDCs), regulatory T cells, myeloid-derived suppressor cells, and CD8(+) regulatory T cells. In contrast, high-dose LPS (10 µg/mouse) reduced lung tumor burden and was associated with a greater influx of pDCs, as well as a stronger Th1 and Th17 polarization. Depletion of pDCs during low-dose LPS administration resulted in a decreased lung tumor burden. Depletion of pDCs during high-dose LPS treatment resulted in an increased tumor burden. The dichotomy in LPS effects was due to the phenotype of pDCs, which were immunosuppressive after the low-dose LPS, and Th1- and T cytotoxic-polarizing cells after the high-dose LPS. Adoptive transfer of T cells into nude mice demonstrated that CD8(+) T cells were responsible for pDC recruitment following low-dose LPS administration, whereas CD4(+) T cells were required for pDC influx after the high-dose LPS. In conclusion, our data suggest differential effects of low-dose versus high-dose LPS on pDC phenotype and tumor progression or regression in the lungs of mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lipopolysaccharides/pharmacology , Lung Neoplasms/pathology , Melanoma, Experimental/pathology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Dendritic Cells/drug effects , Dendritic Cells/pathology , Disease Progression , Dose-Response Relationship, Immunologic , Female , Lipopolysaccharides/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Mice , Mice, Nude , Spleen/drug effects , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/transplantation , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/pathology
18.
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
19.
Front Neurosci ; 18: 1393293, 2024.
Article in English | MEDLINE | ID: mdl-38770241

ABSTRACT

While recent advances in diagnostics and therapeutics offer promising new approaches for Alzheimer's disease (AD) diagnosis and treatment, there is still an unmet need for an effective remedy, suggesting new avenues of research are required. Besides many plausible etiologies for AD pathogenesis, mounting evidence supports a possible role for microbial infections. Various microbes have been identified in the postmortem brain tissues of human AD patients. Among bacterial pathogens in AD, Chlamydia pneumoniae (Cp) has been well characterized in human AD brains and is a leading candidate for an infectious involvement. However, no definitive studies have been performed proving or disproving Cp's role as a causative or accelerating agent in AD pathology and cognitive decline. In this review, we discuss recent updates for the role of Cp in human AD brains as well as experimental models of AD. Furthermore, based on the current literature, we have compiled a list of potential mechanistic pathways which may connect Cp with AD pathology.

20.
Front Immunol ; 15: 1411979, 2024.
Article in English | MEDLINE | ID: mdl-38989288

ABSTRACT

Background: Kawasaki disease (KD), an acute febrile illness and systemic vasculitis, is the leading cause of acquired heart disease in children in industrialized countries. KD leads to the development of coronary artery aneurysms (CAA) in affected children, which may persist for months and even years after the acute phase of the disease. There is an unmet need to characterize the immune and pathological mechanisms of the long-term complications of KD. Methods: We examined cardiovascular complications in the Lactobacillus casei cell wall extract (LCWE) mouse model of KD-like vasculitis over 4 months. The long-term immune, pathological, and functional changes occurring in cardiovascular lesions were characterized by histological examination, flow cytometric analysis, immunofluorescent staining of cardiovascular tissues, and transthoracic echocardiogram. Results: CAA and abdominal aorta dilations were detected up to 16 weeks following LCWE injection and initiation of acute vasculitis. We observed alterations in the composition of circulating immune cell profiles, such as increased monocyte frequencies in the acute phase of the disease and higher counts of neutrophils. We determined a positive correlation between circulating neutrophil and inflammatory monocyte counts and the severity of cardiovascular lesions early after LCWE injection. LCWE-induced KD-like vasculitis was associated with myocarditis and myocardial dysfunction, characterized by diminished ejection fraction and left ventricular remodeling, which worsened over time. We observed extensive fibrosis within the inflamed cardiac tissue early in the disease and myocardial fibrosis in later stages. Conclusion: Our findings indicate that increased circulating neutrophil counts in the acute phase are a reliable predictor of cardiovascular inflammation severity in LCWE-injected mice. Furthermore, long-term cardiac complications stemming from inflammatory cell infiltrations in the aortic root and coronary arteries, myocardial dysfunction, and myocardial fibrosis persist over long periods and are still detected up to 16 weeks after LCWE injection.


Subject(s)
Cell Wall , Disease Models, Animal , Fibrosis , Lacticaseibacillus casei , Mucocutaneous Lymph Node Syndrome , Vasculitis , Animals , Mice , Cell Wall/immunology , Vasculitis/immunology , Vasculitis/etiology , Vasculitis/pathology , Mucocutaneous Lymph Node Syndrome/immunology , Mucocutaneous Lymph Node Syndrome/complications , Male , Myocarditis/etiology , Myocarditis/pathology , Myocarditis/immunology , Inflammation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL