Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 294(6): 1997-2008, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30523158

ABSTRACT

Toll-like receptors (TLRs) are pathogen-recognition receptors that trigger the innate immune response. Recent reports have identified accessory proteins that provide essential support to TLR function through ligand delivery and receptor trafficking. Herein, we introduce leucine-rich repeats (LRRs) and calponin homology containing 4 (Lrch4) as a novel TLR accessory protein. Lrch4 is a membrane protein with nine LRRs in its predicted ectodomain. It is widely expressed across murine tissues and has two expression variants that are both regulated by lipopolysaccharide (LPS). Predictive modeling indicates that Lrch4 LRRs conform to the horseshoe-shaped structure typical of LRRs in pathogen-recognition receptors and that the best structural match in the protein database is to the variable lymphocyte receptor of the jawless vertebrate hagfish. Silencing Lrch4 attenuates cytokine induction by LPS and multiple other TLR ligands and dampens the in vivo innate immune response. Lrch4 promotes proper docking of LPS in lipid raft membrane microdomains. We provide evidence that this is through regulation of lipid rafts as Lrch4 silencing reduces cell surface gangliosides, a metric of raft abundance, as well as expression and surface display of CD14, a raft-resident LPS co-receptor. Taken together, we identify Lrch4 as a broad-spanning regulator of the innate immune response and a potential molecular target in inflammatory disease.


Subject(s)
Gene Expression Regulation , Immunity, Innate , Toll-Like Receptors , Animals , Gangliosides/metabolism , Leucine , Ligands , Lipopolysaccharide Receptors , Lipopolysaccharides/metabolism , Membrane Microdomains/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Protein Conformation , Protein Domains
2.
J Allergy Clin Immunol ; 134(1): 127-34, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24655576

ABSTRACT

BACKGROUND: The genetic determinants of the human innate immune response are poorly understood. Apolipoprotein (Apo) E, a lipid-trafficking protein that affects inflammation, has well-described wild-type (ε3) and disease-associated (ε2 and ε4) alleles, but its connection to human innate immunity is undefined. OBJECTIVE: We sought to define the relationship of APOε4 to the human innate immune response. METHODS: We evaluated APOε4 in several functional models of the human innate immune response, including intravenous LPS challenge in human subjects, and assessed APOε4 association to organ injury in patients with severe sepsis, a disease driven by dysregulated innate immunity. RESULTS: Whole blood from healthy APOε3/APOε4 volunteers induced higher cytokine levels on ex vivo stimulation with Toll-like receptor (TLR) 2, TLR4, or TLR5 ligands than blood from APOε3/APOε3 patients, whereas TLR7/8 responses were similar. This was associated with increased lipid rafts in APOε3/APOε4 monocytes. By contrast, APOε3/APOε3 and APOε3/APOε4 serum neutralized LPS equivalently and supported similar LPS responses in Apoe-deficient macrophages, arguing against a differential role for secretory APOE4 protein. After intravenous LPS, APOε3/APOε4 patients had higher hyperthermia and plasma TNF-α levels and earlier plasma IL-6 than APOε3/APOε3 patients. APOE4-targeted replacement mice displayed enhanced hypothermia, plasma cytokines, and hepatic injury and altered splenic lymphocyte apoptosis after systemic LPS compared with APOE3 counterparts. In a cohort of 828 patients with severe sepsis, APOε4 was associated with increased coagulation system failure among European American patients. CONCLUSIONS: APOε4 is a determinant of the human innate immune response to multiple TLR ligands and associates with altered patterns of organ injury in human sepsis.


Subject(s)
Apolipoprotein E4/immunology , Immunity, Innate , Sepsis/immunology , Adult , Animals , Apolipoprotein E3/genetics , Apolipoprotein E3/immunology , Apolipoprotein E4/genetics , Cells, Cultured , Gene Expression , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Ligands , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , Sepsis/genetics , Sepsis/pathology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
3.
J Exp Med ; 210(5): 891-904, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23630228

ABSTRACT

Cancer and infection are predominant causes of human mortality and derive, respectively, from inadequate genomic and host defenses against environmental agents. The transcription factor p53 plays a central role in human tumor suppression. Despite its expression in immune cells and broad responsiveness to stressors, it is virtually unknown whether p53 regulates host defense against infection. We report that the lungs of naive p53(-/-) mice display genome-wide induction of NF-κB response element-enriched proinflammatory genes, suggestive of type 1 immune priming. p53-null and p53 inhibitor-treated mice clear Gram-negative and -positive bacteria more effectively than controls after intrapulmonary infection. This is caused, at least in part, by cytokines produced by an expanded population of apoptosis-resistant, TLR-hyperresponsive alveolar macrophages that enhance airway neutrophilia. p53(-/-) neutrophils, in turn, display heightened phagocytosis, Nox-dependent oxidant generation, degranulation, and bacterial killing. p53 inhibition boosts bacterial killing by mouse neutrophils and oxidant generation by human neutrophils. Despite enhanced bacterial clearance, infected p53(-/-) mice suffer increased mortality associated with aggravated lung injury. p53 thus modulates host defense through regulating microbicidal function and fate of phagocytes, revealing a fundamental link between defense of genome and host during environmental insult.


Subject(s)
Cell Lineage/immunology , Host-Pathogen Interactions/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Tumor Suppressor Protein p53/metabolism , Animals , Anti-Infective Agents/pharmacology , Cell Death/drug effects , Cell Lineage/drug effects , Cell Movement/drug effects , Cell Movement/immunology , Cytokines/metabolism , Female , Gene Deletion , Genome/genetics , Host-Pathogen Interactions/drug effects , Humans , Inflammation/genetics , Inflammation/immunology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/immunology , Leukocyte Count , Lung/drug effects , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophages/metabolism , Male , Mice , NF-kappa B/metabolism , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Nitric Oxide/biosynthesis , Pneumonia, Bacterial/pathology , Survival Analysis , Toll-Like Receptors/metabolism , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/deficiency
4.
J Immunol ; 188(11): 5327-36, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22539789

ABSTRACT

Mice with genetic deletion of the cholesterol transporter ATP binding cassette G1 (ABCG1) have pulmonary lipidosis and enhanced innate immune responses in the airway. Whether ABCG1 regulates adaptive immune responses to the environment is unknown. To this end, Abcg1(+/+) and Abcg1(-/-) mice were sensitized to OVA via the airway using low-dose LPS as an adjuvant, and then challenged with OVA aerosol. Naive Abcg1(-/-) mice displayed increased B cells, CD4(+) T cells, CD8(+) T cells, and dendritic cells (DCs) in lung and lung-draining mediastinal lymph nodes, with lung CD11b(+) DCs displaying increased CD80 and CD86. Upon allergen sensitization and challenge, the Abcg1(-/-) airway, compared with Abcg1(+/+), displayed reduced Th2 responses (IL-4, IL-5, eosinophils), increased neutrophils and IL-17, but equivalent airway hyperresponsiveness. Reduced Th2 responses were also found using standard i.p. OVA sensitization with aluminum hydroxide adjuvant. Mediastinal lymph nodes from airway-sensitized Abcg1(-/-) mice produced reduced IL-5 upon ex vivo OVA challenge. Abcg1(-/-) CD4(+) T cells displayed normal ex vivo differentiation, whereas Abcg1(-/-) DCs were found paradoxically to promote Th2 polarization. Th17 cells, IL-17(+) γδT cells, and IL-17(+) neutrophils were all increased in Abcg1(-/-) lungs, suggesting Th17 and non-Th17 sources of IL-17 excess. Neutralization of IL-17 prior to challenge normalized eosinophils and reduced neutrophilia in the Abcg1(-/-) airway. We conclude that Abcg1(-/-) mice display IL-17-mediated suppression of eosinophilia and enhancement of neutrophilia in the airway following allergen sensitization and challenge. These findings identify ABCG1 as a novel integrator of cholesterol homeostasis and adaptive immune programs.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Adaptive Immunity/genetics , Eosinophilia/immunology , Interleukin-17/physiology , Lipoproteins/deficiency , Lipoproteins/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/physiology , Animals , Disease Models, Animal , Eosinophilia/genetics , Eosinophilia/pathology , Gene Knockdown Techniques , Lipoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology
5.
Mol Cell Biol ; 30(19): 4712-21, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20647536

ABSTRACT

Macrophage activation and infiltration into resident tissues is known to mediate local inflammation and is a hallmark feature of metabolic syndrome. Members of the sirtuin family of proteins regulate numerous physiological processes, including those involved in nutrient regulation and the promotion of longevity. However, the important role that SIRT1, the leading sirtuin family member, plays in immune response remains unclear. In this study, we demonstrate that SIRT1 modulates the acetylation status of the RelA/p65 subunit of NF-κB and thus plays a pivotal role in regulating the inflammatory, immune, and apoptotic responses in mammals. Using a myeloid cell-specific SIRT1 knockout (Mac-SIRT1 KO) mouse model, we show that ablation of SIRT1 in macrophages renders NF-κB hyperacetylated, resulting in increased transcriptional activation of proinflammatory target genes. Consistent with increased proinflammatory gene expression, Mac-SIRT1 KO mice challenged with a high-fat diet display high levels of activated macrophages in liver and adipose tissue, predisposing the animals to development of systemic insulin resistance and metabolic derangement. In summary, we report that SIRT1, in macrophages, functions to inhibit NF-κB-mediated transcription, implying that myeloid cell-specific modulation of this sirtuin may be beneficial in the treatment of inflammation and its associated diseases.


Subject(s)
Inflammation/metabolism , Signal Transduction , Sirtuin 1/metabolism , Transcription Factor RelA/metabolism , Acetylation , Animals , Blotting, Western , Cells, Cultured , Cytokines/metabolism , Dietary Fats/administration & dosage , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Histone Acetyltransferases/metabolism , Inflammation/genetics , L Cells , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , RNA Interference , Sirtuin 1/genetics , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/pharmacology
6.
J Immunol ; 185(3): 1660-9, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20581153

ABSTRACT

Dyslipidemia influences innate immune responses in the bloodstream, but whether and how pulmonary innate immunity is sensitive to circulating lipoproteins is largely unknown. To define whether dyslipidemia impacts responses to bacteria in the airspace and, if so, whether differently from its effects in other tissues, airspace, bloodstream, and i.p. responses to LPS and Klebsiella pneumoniae were investigated using murine models of dyslipidemia. Dyslipidemia reduced neutrophil (PMN) recruitment to the airspace in response to LPS and K. pneumoniae by impairing both chemokine induction in the airspace and PMN chemotaxis, thereby compromising pulmonary bacterial clearance. Paradoxically, bacteria were cleared more effectively from the bloodstream during dyslipidemia. This enhanced systemic response was due, at least in part, to basal circulating neutrophilia and basal TLR4/MyD88-dependent serum cytokine induction and enhanced serum cytokine responses to systemically administered TLR ligands. Dyslipidemia did not globally impair PMN transvascular trafficking to, and host defense within all loci, because neutrophilia, cytokine induction, and bacterial clearance were enhanced within the infected peritoneum. Peritoneal macrophages from dyslipidemic animals were primed for more robust TLR responses, reflecting increased lipid rafts and increased TLR4 expression, whereas macrophages from the airspace, in which cholesterol was maintained constant during dyslipidemia, had normal responses and rafts. Dyslipidemia thus imparts opposing effects upon intra- and extrapulmonary host defense by inducing tissue-divergent TLR response phenotypes and dysregulating airspace/blood compartmental levels of PMNs and cytokines. We propose that the airspace is a "privileged" site, thereby uniquely sensitive to dyslipidemia.


Subject(s)
Dyslipidemias/immunology , Dyslipidemias/metabolism , Immunity, Innate , Klebsiella Infections/immunology , Pneumonia, Bacterial/immunology , Toll-Like Receptors/biosynthesis , Animals , Cell Line , Cells, Cultured , Cytokines/biosynthesis , Dyslipidemias/pathology , Female , Immunophenotyping , Klebsiella Infections/microbiology , Klebsiella Infections/pathology , Klebsiella pneumoniae/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Toll-Like Receptors/blood
7.
Am J Respir Crit Care Med ; 182(3): 404-12, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20395559

ABSTRACT

RATIONALE: Mice with genetic deletion of the cholesterol efflux transporter, ATP-binding cassette (ABC) G1, have pulmonary lipidosis and chronic pulmonary inflammation. Whether ABCG1 regulates host defense is unknown. OBJECTIVES: To determine whether ABCG1 regulates pulmonary innate immunity and host defense, and to investigate the underlying molecular/cellular mechanisms. METHODS: Abcg1(+/+) and Abcg1(-/-) mice were challenged with intrapulmonary lipopolysaccharide (LPS) or Klebsiella pneumoniae, intravenous K. pneumoniae, or intraperitoneal LPS. Phenotypic responses were profiled. Bone marrow chimeras and in vitro assays were used to differentiate and characterize the role of hematopoietic versus nonhematopoietic ABCG1 in host defense. MEASUREMENTS AND MAIN RESULTS: Unexposed Abcg1(-/-) mice had normal numbers of circulating neutrophils, but increased neutrophil recruitment to the airspace and lung parenchyma, and increased airspace cytokines and chemokines in the steady state. After intrapulmonary LPS or K. pneumoniae, Abcg1(-/-) mice displayed exaggerated further neutrophil recruitment to and degranulation in the airspace, and elevated airspace cytokine/chemokine induction. Alveolar macrophage ABCG1 was critical, as ABCG1 deficiency in hematopoietic cells was sufficient to enhance responses in vivo, and Abcg1(-/-) alveolar macrophages adopted a "foam cell" phenotype, and were hyperresponsive ex vivo. Pulmonary compartmentalization and clearance of K. pneumoniae were increased in Abcg1(-/-) mice, indicating enhanced host defense. By contrast, Abcg1(+/+) and Abcg1(-/-) mice had equivalent responses to intravenous K. pneumoniae and intraperitoneal LPS, suggesting that ABCG1 regulates innate immunity in a tissue-selective manner. CONCLUSIONS: Abcg1(-/-) mice have an enhanced pulmonary host defense response driven predominantly by hematopoietic cells.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Immunity, Innate , Lipoproteins/physiology , Lung/immunology , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Animals , Bronchoalveolar Lavage Fluid/cytology , Cytokines/metabolism , Gene Deletion , Klebsiella pneumoniae , Leukocyte Count , Leukocytes/metabolism , Lipopolysaccharides/administration & dosage , Lipoproteins/genetics , Lung/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Neutrophil Activation , Neutrophils/metabolism
8.
J Allergy Clin Immunol ; 124(5): 967-74.e1-15, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19800678

ABSTRACT

BACKGROUND: Cholesterol exerts complex effects on inflammation. There has been little investigation of whether serum cholesterol is associated with asthma, an inflammatory airways disease with great public health impact. OBJECTIVE: To determine relationships between levels of 3 serum cholesterol measures (total cholesterol [TC], high-density lipoprotein cholesterol [HDL-C], and non-HDL-C) and asthma/wheeze in a sample representative of the US population. METHODS: Cross-sectional study of 7005 participants age >or=6 years from the 2005 to 2006 National Health and Nutrition Examination Survey. RESULTS: Serum TC and non-HDL-C were lower in patients with current asthma than in subjects without current asthma in the overall population (TC, 188.5 vs 192.2 mg/dL; non-HDL-C, 133.9 vs 137.7 mg/dL; P < .05 for both), whereas HDL-C was not different. Adjusted odds ratios (ORs) from multivariate logistic regression per 1-SD increase of TC and non-HDL-C for current asthma were 0.92 (95% CI, 0.86-0.98) and 0.91 (95% CI, 0.85-0.98), respectively. On racial/ethnic stratification, these relationships reflect marked reductions unique to Mexican Americans (MAs; TC, 171.4 vs 189.3 mg/dL; P < .001; OR, 0.62; 95% CI, 0.48-0.80; non-HDL-C, 119.8 vs 137.9 mg/dL; P < .001; OR, 0.62; 95% CI, 0.48-0.79). Among MAs, the adjusted OR for wheeze requiring medical attention was 0.57 (95% CI, 0.43-0.75) for TC and 0.53 (95% CI, 0.33-0.85) for non-HDL-C. Relationships between cholesterol and asthma/wheeze were independent of body mass index and serum C-reactive protein, and similar between atopic and nonatopic participants. CONCLUSION: Serum TC and non-HDL-C are inversely related to asthma in the US population, chiefly reflecting a relationship among MAs.


Subject(s)
Asthma/epidemiology , Cholesterol/blood , Respiratory Sounds , Adult , Asthma/blood , Body Mass Index , C-Reactive Protein/analysis , Cross-Sectional Studies , Female , Humans , Logistic Models , Male , Multivariate Analysis , United States/epidemiology
9.
Immunol Lett ; 102(2): 202-14, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16242782

ABSTRACT

Group B streptococcus (GBS), a capsulated gram-positive bacterium, is a major cause of newborn infections. Although the innate immune receptor Toll-like receptor (TLR) 2 has been shown to primarily recognize gram-positive bacterial products, the production of TNF by macrophages treated with heat-killed GBS (HK-GBS) does not depend on TLR2. In this report, we have characterized HK-GBS-induced activation of macrophages derived from wildtype and TLR2-deficient mice. Microarray analysis demonstrated that HK-GBS activation of macrophages induces both TLR2-independent and -dependent signals. While the expression of a major fraction of genes in macrophages induced by HK-GBS does not depend on TLR2, induction of several important molecules involved in host innate immunity such as IL-6, IL-1beta, and lipocalin 2 is severely impaired in the absence of TLR2 signaling. Furthermore, we show that HK-GBS utilizes centrifugation sensitive components to induce rapid activation of TLR2(-/-) macrophages and that HK-GBS-induced activation of macrophages is not mediated through its genomic DNA. Together, our results demonstrate that HK-GBS induces TLR2-dependent antimicrobial gene activation and provide further understanding of the molecular basis of host innate response to GBS infection.


Subject(s)
Inflammation/immunology , Streptococcus agalactiae/immunology , Toll-Like Receptor 2/physiology , Animals , Hot Temperature , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 2/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation
10.
J Immunol ; 172(4): 2416-23, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14764712

ABSTRACT

In these experiments, we investigated the role of calcium as a second messenger in the apoptotic activation of cytosolic phospholipase A(2) (cPLA(2)). As our model, we used a murine fibroblast cell line (C3HA) that was induced to undergo apoptosis by a combination of TNF and cycloheximide. Using fura 2 Ca(2+) imaging, we found strong evidence for an intracellular calcium response after 1 h of treatment, which correlated with the onset of phosphatidylserine externalization, but preceded effector procaspase processing by several hours. The response was strongest in the perinuclear region, where mean levels rose 83% (144 +/- 14 nM in untreated cells vs 264 +/- 39 nM in treated), while cells displaying morphological evidence of apoptosis had the highest levels of calcium (250-1000 nM). Verapamil blocked this response, indicating an extracellular source for the calcium. Fluorescence microscopy revealed a pattern of nuclear translocation of cPLA(2) during apoptosis, which was also blocked by verapamil, indicating an important role for calcium in this process. In addition, we found that verapamil prevented the release of [(3)H]arachidonic acid from C3HA cells induced to undergo apoptosis by the chemotherapeutic agents vinblastine, melphalan, and cis-platinum. Together, these data suggest that calcium is important for cPLA(2) activation by diverse apoptotic stimuli.


Subject(s)
Apoptosis/drug effects , Apoptosis/immunology , Calcium/physiology , Cell Nucleus/enzymology , Cycloheximide/pharmacology , Phospholipases A/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/immunology , Animals , Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/metabolism , Calcium/metabolism , Caspases/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/immunology , Cytosol/drug effects , Cytosol/enzymology , Cytosol/immunology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Hydrolysis/drug effects , Intracellular Fluid/drug effects , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Mice , Mice, Inbred C3H , Phosphatidylserines/metabolism , Phospholipases A2 , Tritium , Up-Regulation/drug effects , Up-Regulation/immunology , Verapamil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...