Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mediators Inflamm ; 2018: 6808934, 2018.
Article in English | MEDLINE | ID: mdl-30532653

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is an important regulator of virus-induced antiviral interferons (IFNs) and proinflammatory cytokines. It requires interaction with an adaptor molecule, mitochondrial antiviral-signaling protein (MAVS), to activate downstream signaling pathways. To elucidate the mechanism(s) by which RIG-I-dependent recognition of IAV infection in vivo triggers innate immune responses, we infected mutant mice lacking RIG-I or MAVS with influenza A virus (IAV) and measured their innate immune responses. As has previously been demonstrated with isolated deletion of the virus recognition receptors TLR3, TLR7, and NOD2, RIG-I or MAVS knockout (KO) did not result in higher mortality and did not reduce IAV-induced cytokine responses in mice. Infected RIG-I KO animals displayed similar lung inflammation profiles as did WT mice, in terms of the protein concentration, total cell count, and inflammatory cell composition in the bronchoalveolar lavage fluid. RNA-Seq results demonstrated that all types of mice exhibited equivalent antiviral and inflammatory gene responses following IAV infection. Together, the results indicated that although RIG-I is important in innate cytokine responses in vitro, individual deletion of the genes encoding RIG-I or MAVS did not change survival or innate responses in vivo after IAV infection in mice.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD Box Protein 58/metabolism , Influenza A virus/pathogenicity , Orthomyxoviridae Infections/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bronchoalveolar Lavage , DEAD Box Protein 58/genetics , Humans , Immunoassay , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Signal Transduction/physiology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism
2.
Microb Pathog ; 121: 9-21, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29704667

ABSTRACT

The lung is the entry site for Bacillus anthracis in inhalation anthrax, the most deadly form of the disease. Spores must escape through the alveolar epithelial cell (AEC) barrier and migrate to regional lymph nodes, germinate and enter the circulatory system to cause disease. Several mechanisms to explain alveolar escape have been postulated, and all these tacitly involve the AEC barrier. In this study, we incorporate our primary human type I AEC model, microarray and gene enrichment analysis, qRT-PCR, multiplex ELISA, and neutrophil and monocyte chemotaxis assays to study the response of AEC to B. anthracis, (Sterne) spores at 4 and 24 h post-exposure. Spore exposure altered gene expression in AEC after 4 and 24 h and differentially expressed genes (±1.3 fold, p ≤ 0.05) included CCL4/MIP-1ß (4 h), CXCL8/IL-8 (4 and 24 h) and CXCL5/ENA-78 (24 h). Gene enrichment analysis revealed that pathways involving cytokine or chemokine activity, receptor binding, and innate immune responses to infection were prominent. Microarray results were confirmed by qRT-PCR and multiplex ELISA assays. Chemotaxis assays demonstrated that spores induced the release of biologically active neutrophil and monocyte chemokines, and that CXCL8/IL-8 was the major neutrophil chemokine. The small or sub-chemotactic doses of CXCL5/ENA-78, CXCL2/GROß and CCL20/MIP-3α may contribute to chemotaxis by priming effects. These data provide the first whole transcriptomic description of the human type I AEC initial response to B. anthracis spore exposure. Taken together, our findings contribute to an increased understanding of the role of AEC in the pathogenesis of inhalational anthrax.


Subject(s)
Alveolar Epithelial Cells/microbiology , Bacillus anthracis/pathogenicity , Chemokines/metabolism , Gene Expression Profiling , Spores, Bacterial/pathogenicity , Anthrax/genetics , Anthrax/metabolism , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Chemokine CXCL5/genetics , Chemokine CXCL5/metabolism , Chemokines/genetics , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Monocytes/metabolism , Monocytes/microbiology , Neutrophils/metabolism , Neutrophils/microbiology , Platelet Factor 4/genetics , Platelet Factor 4/metabolism , Respiratory Tract Infections/genetics , Respiratory Tract Infections/metabolism , Up-Regulation
3.
Respir Res ; 18(1): 166, 2017 09 02.
Article in English | MEDLINE | ID: mdl-28865477

ABSTRACT

BACKGROUND: Retinoic acid-inducible gene I (RIG-I) is an important regulator of virus-induced antiviral interferons (IFNs) and proinflammatory cytokines which participate in clearing viral infections. Cigarette smoke (CS) exposure increases the frequency and severity of respiratory tract infections. METHODS: We generated a RIG-I transgenic (TG) mouse strain that expresses the RIG-I gene product under the control of the human lung specific surfactant protein C promoter. We compared the mortality and host immune responses of RIG-I TG mice and their litter-matched wild type (WT) mice following challenge with influenza A virus (IAV). RESULTS: RIG-I overexpression increased survival of IAV-infected mice. CS exposure increased mortality in WT mice infected with IAV. Remarkably, the effect of RIG-I overexpression on survival during IAV infection was enhanced in CS-exposed animals. CS-exposed IAV-infected WT mice had a suppressed innate response profile in the lung compared to sham-exposed IAV-infected WT mice in terms of the protein concentration, total cell count and inflammatory cell composition in the bronchoalveolar lavage fluid. RIG-I overexpression restored the innate immune response in CS-exposed mice to that seen in sham-exposed WT mice during IAV infection, and is likely responsible for enhanced survival in RIG-I TG mice as restoration preceded death of the animals. CONCLUSIONS: Our results demonstrate that RIG-I overexpression in mice is protective for CS enhanced susceptibility of smokers to influenza infection, and that CS mediated RIG-I suppression may be partially responsible for the increased morbidity and mortality of the mice exposed to IAV. Thus, optimizing the RIG-I response may be an important treatment strategy for CS-enhanced lung infections, particularly those due to IAV.


Subject(s)
DEAD Box Protein 58/biosynthesis , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/mortality , Smoking/metabolism , Smoking/mortality , Animals , DEAD Box Protein 58/genetics , Dogs , Gene Expression , Humans , Inhalation Exposure/adverse effects , Madin Darby Canine Kidney Cells , Mice , Mice, Transgenic , Mortality/trends , Smoking/adverse effects , Tobacco Smoke Pollution/adverse effects
4.
J Immunol ; 198(3): 1183-1201, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28031342

ABSTRACT

The respiratory system is a complex network of many cell types, including subsets of macrophages and dendritic cells that work together to maintain steady-state respiration. Owing to limitations in acquiring cells from healthy human lung, these subsets remain poorly characterized transcriptionally and phenotypically. We set out to systematically identify these subsets in human airways by developing a schema of isolating large numbers of cells by whole-lung bronchoalveolar lavage. Six subsets of phagocytic APC (HLA-DR+) were consistently observed. Aside from alveolar macrophages, subsets of Langerin+, BDCA1-CD14+, BDCA1+CD14+, BDCA1+CD14-, and BDCA1-CD14- cells were identified. These subsets varied in their ability to internalize Escherichia coli, Staphylococcus aureus, and Bacillus anthracis particles. All subsets were more efficient at internalizing S. aureus and B. anthracis compared with E. coli Alveolar macrophages and CD14+ cells were overall more efficient at particle internalization compared with the four other populations. Subsets were further separated into two groups based on their inherent capacities to upregulate surface CD83, CD86, and CCR7 expression levels. Whole-genome transcriptional profiling revealed a clade of "true dendritic cells" consisting of Langerin+, BDCA1+CD14+, and BDCA1+CD14- cells. The dendritic cell clade was distinct from a macrophage/monocyte clade, as supported by higher mRNA expression levels of several dendritic cell-associated genes, including CD1, FLT3, CX3CR1, and CCR6 Each clade, and each member of both clades, was discerned by specific upregulated genes, which can serve as markers for future studies in healthy and diseased states.


Subject(s)
Dendritic Cells/physiology , Lung/immunology , Macrophages, Alveolar/physiology , Macrophages/physiology , Adult , Aged , Antigens, CD/analysis , Antigens, CD1/analysis , B7-2 Antigen/analysis , Dendritic Cells/classification , Gene Expression Profiling , Glycoproteins/analysis , Humans , Immunoglobulins/analysis , Lipopolysaccharide Receptors/analysis , Lung/microbiology , Macrophages/classification , Membrane Glycoproteins/analysis , Middle Aged , CD83 Antigen
5.
Microbes Infect ; 18(10): 615-626, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27320392

ABSTRACT

The lung is the entry site for Bacillus anthracis in inhalation anthrax, the most deadly form of the disease. Spores escape from the alveolus to regional lymph nodes, germinate and enter the circulatory system to cause disease. The roles of carrier cells and the effects of B. anthracis toxins in this process are unclear. We used a human lung organ culture model to measure spore uptake by antigen presenting cells (APC) and alveolar epithelial cells (AEC), spore partitioning between these cells, and the effects of B. anthracis lethal toxin and protective antigen. We repeated the study in a human A549 alveolar epithelial cell model. Most spores remained unassociated with cells, but the majority of cell-associated spores were in AEC, not in APC. Spore movement was not dependent on internalization, although the location of internalized spores changed in both cell types. Spores also internalized in a non-uniform pattern. Toxins affected neither transit of the spores nor the partitioning of spores into AEC and APC. Our results support a model of spore escape from the alveolus that involves spore clustering with transient passage through intact AEC. However, subsequent transport of spores by APC from the lung to the lymph nodes may occur.


Subject(s)
Anthrax/pathology , Antigens, Bacterial/metabolism , Bacillus anthracis/pathogenicity , Bacterial Toxins/metabolism , Lung/microbiology , Lymph Nodes/microbiology , Movement , Spores, Bacterial/pathogenicity , Antigen-Presenting Cells/microbiology , Blood/microbiology , Cell Line , Epithelial Cells/microbiology , Humans , Models, Theoretical , Organ Culture Techniques
6.
Virology ; 482: 181-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25880109

ABSTRACT

Pattern recognition receptors, such as retinoic acid-inducible protein I (RIG-I), Toll-like receptors 3 and 7 (TLR3 and 7), and nucleotide-binding oligomerization domain containing protein 2 (NOD2), play important roles in the recognition of influenza A virus (IAV), but their role in interferon (IFN) induction is still unclear, particularly in human lung. We investigated IFN induction by IAV in the A549 cell line as well as in primary human alveolar epithelial cells (AEC). TLR3/7, NOD2, RIG-I, and IFN expression levels were measured by qRT-PCR and ELISA in cells infected with IAV PR8. We found that TLR7 and NOD2 were not involved in IFN induction by IAV in these cells. Neither RIG-I nor TLR3 siRNA alone completely blocked IFN induction. However, double knockdown of RIG-I and TLR3 completely inhibited IFN induction by influenza. Thus, signaling through both RIG-I and TLR3 is important for IFN induction by IAV in human lung AEC.


Subject(s)
DEAD-box RNA Helicases/metabolism , Epithelial Cells/immunology , Epithelial Cells/virology , Influenza A Virus, H1N1 Subtype/immunology , Interferons/metabolism , Toll-Like Receptor 3/metabolism , Cells, Cultured , DEAD Box Protein 58 , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Real-Time Polymerase Chain Reaction , Receptors, Immunologic
7.
Am J Physiol Lung Cell Mol Physiol ; 307(11): L848-58, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25260755

ABSTRACT

Cigarette smoke (CS) exposure increases the frequency and severity of respiratory tract infections. Despite this association, the mechanisms underlying the increased susceptibility to respiratory virus infection are poorly understood. Retinoic acid-inducible gene I (RIG-I) is an important regulator of influenza virus-induced expression of antiviral cytokines, mainly interferons (IFNs), which are necessary to clear viral infections. In this study, we compared the innate cytokine responses of two mouse CS exposure models following a challenge with influenza A virus (IAV): 1) exposure of the mice to cigarette smoke extract (CSE) intratracheally and 2) exposure of the mice to CS in a whole body exposure chamber. Both intratracheal CSE treatment and whole body CS exposure caused antiviral immunosuppression in these mice, and both CS exposure methods inhibited RIG-I induction. CS attenuated influenza-induced antiviral IFNs and IP-10 expression in vivo. However, we did not find that CS inhibited induction of the proinflammatory cytokines IL-6 and TNF-α, whose expression was induced by IAV. Interestingly, IAV infection also increased Toll-like receptor 3 (TLR3) expression in mouse lung, but CS exposure did not impact TLR3 induction in these mice. Together, the results support our previous finding in a human lung organ culture model that the suppression of RIG-I induction and antiviral cytokine responses by CS are likely important in the enhanced susceptibility of smokers to influenza infection in the lung.


Subject(s)
DEAD-box RNA Helicases/biosynthesis , Immunity, Innate/drug effects , Influenza A Virus, H1N1 Subtype/immunology , Nicotiana/adverse effects , Orthomyxoviridae Infections/immunology , Smoke/adverse effects , Animals , Bronchoalveolar Lavage Fluid/immunology , Chemokine CXCL10/biosynthesis , DEAD Box Protein 58 , Female , Immune Tolerance/drug effects , Immune Tolerance/immunology , Immunity, Innate/immunology , Interferon-beta/biosynthesis , Interleukin-6/biosynthesis , Lung/immunology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/prevention & control , Smoking/adverse effects , Toll-Like Receptor 3/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
8.
J Gen Virol ; 91(Pt 5): 1155-63, 2010 May.
Article in English | MEDLINE | ID: mdl-20071488

ABSTRACT

Adenovirus (Ad) type 7 can cause severe infection, including pneumonia, in military recruits and children. The initial inflammation is a neutrophilic interstitial infiltration with neutrophilic alveolitis. Subsequently, monocytes become evident and, finally, there is a predominantly lymphocytic infiltrate. We have established that Ad7 infection of epithelial cells stimulates release of the neutrophil chemotaxin interleukin (IL)-8, and have extended these studies to a human lung tissue model. Here, we studied cytokine responses to Ad7 in human alveolar macrophages (HAM) and our human lung tissue model. Both ELISA and RNase-protection assay (RPA) data demonstrated that, upon Ad7 infection, IP-10 and MIP-1alpha/beta are released from HAM. IP-10 and MIP-1alpha/beta protein levels were induced 2- and 3-fold, respectively, in HAM 24 h after Ad7 infection. We then investigated induction of specific cytokines in human lung tissue by RPA and ELISA. The results showed that IL-8 and IL-6 were induced 8 h after infection and, by 24 h, levels of IL-8, IL-6, MIP-1alpha/beta and MCP-1 were all increased. IP-10, a monocyte and lymphocyte chemokine, was also induced 30-fold, but only 24 h after infection. Immunohistochemistry staining confirmed that IL-8 was only released from the epithelial cells of lung slices and not from macrophages. IP-10 was secreted from both macrophages and epithelial cells. Moreover, full induction of IP-10 is likely to require participation and cooperation of both epithelial cells and macrophages in intact lung. Understanding the cytokine and chemokine induction during Ad7 infection may lead to novel ways to modulate the response to this pathogen.


Subject(s)
Adenoviridae Infections/immunology , Adenoviruses, Human/immunology , Cytokines/metabolism , Immunity, Innate , Lung/immunology , Pneumonia, Viral/immunology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology
9.
Virology ; 396(2): 178-88, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-19913271

ABSTRACT

We studied cytokine responses to influenza virus PR8 (H1N1) and Oklahoma/309/06 (OK/06, H3N2) in a novel human lung tissue model. Exposure of the model to influenza virus rapidly activated the mitogen-activated protein kinase signaling (MAPK) pathways ERK, p38 and JNK. In addition, RNase protection assay demonstrated the induction of several cytokine and chemokine mRNAs by virus. This finding was reflected at the translational level as IL-6, MCP-1, MIP-1 alpha/beta, IL-8 and IP-10 proteins were induced as determined by ELISA. Immunohistochemistry for IP-10 and MIP-1 alpha revealed that alveolar epithelial cells and macrophages were the source of these two cytokines. Taken together, both PR8 and OK/06 cause similar induction of cytokines in human lung, although OK/06 is less effective at inducing the chemokines MCP-1 and IL-8. This human organ culture model should thus provide a relevant platform to study the biological responses of human lung to influenza virus infection.


Subject(s)
Immunity, Innate/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Lung/virology , Chemokines/biosynthesis , Cytokines/biosynthesis , Enzyme Induction/immunology , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/virology , Lung/immunology , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Organ Culture Techniques , Signal Transduction/immunology
10.
J Immunol ; 183(9): 5799-806, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812208

ABSTRACT

The etiologic agent of inhalational anthrax, Bacillus anthracis, produces virulence toxins that are important in the disease pathogenesis. Current studies suggest that mouse and human macrophages are susceptible to immunosuppressive effects of one of the virulence toxins, lethal toxin (LT). Thus a paradigm has emerged that holds that the alveolar macrophage (AM) does not play a significant role in the innate immune response to B. anthracis or defend against the pathogen as it is disabled by LT. This is inconsistent with animal models and autopsy studies that show minimal disease at the alveolar surface. We examined whether AM are immunosuppressed by LT. We found that human AM were relatively resistant to LT-mediated innate immune cytokine suppression, MEK cleavage, and induction of apoptosis as compared with mouse RAW 264.7 macrophages. Mouse AM and murine bone marrow-derived macrophages were also relatively resistant to LT-mediated apoptosis despite intermediate sensitivity to MEK cleavage. The binding component of LT, protective Ag, does not attach to human AM, although it did bind to mouse AM, murine bone marrow-derived macrophages, and RAW 264.7 macrophages. Human AM do not produce significant amounts of the protective Ag receptor anthrax toxin receptor 1 (TEM8/ANTXR1) and anthrax toxin receptor 2 (CMG2/ANTXR2). Thus, mature and differentiated AM are relatively resistant to the effects of LT as compared with mouse RAW 264.7 macrophages. AM resistance to LT may enhance clearance of the pathogen from the alveolar surface and explain why this surface is relatively free of B. anthracis in animal models and autopsy studies.


Subject(s)
Anthrax/immunology , Anthrax/mortality , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Immunity, Innate , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Animals , Anthrax/microbiology , Antigens, Bacterial/toxicity , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/toxicity , Macrophages, Alveolar/enzymology , Mice , Mice, Inbred C57BL , Virulence/immunology
11.
Infect Immun ; 75(8): 3729-38, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17517878

ABSTRACT

Bacillus anthracis, the causative agent of inhalational anthrax, enters a host through the pulmonary system before dissemination. We have previously shown that human alveolar macrophages participate in the initial innate immune response to B. anthracis spores through cell signal-mediated cytokine release. We proposed that the lung epithelia also participate in the innate immune response to this pathogen, and we have developed a human lung slice model to study this process. Exposure of our model to B. anthracis (Sterne) spores rapidly activated the mitogen-activated protein kinase signaling pathways ERK, p38, and JNK. In addition, an RNase protection assay showed induction of mRNA of several cytokines and chemokines. This finding was reflected at the translational level by protein peak increases of 3-, 25-, 9-, 34-, and 5-fold for interleukin-6 (IL-6), tumor necrosis factor alpha, IL-8, macrophage inflammatory protein 1alpha/beta, and monocyte chemoattractant protein 1, respectively, as determined by an enzyme-linked immunosorbent assay. Inhibition of individual pathways by UO126, SP600125, and SB0203580 decreased induction of chemokines and cytokines by spores, but this depended on the pathways inhibited and the cytokines and chemokines induced. Combining all three inhibitors reduced induction of all cytokines and chemokines tested to background levels. An immunohistochemistry analysis of IL-6 and IL-8 revealed that alveolar epithelial cells and macrophages and a few interstitial cells are the source of the cytokines and chemokines. Taken together, these data showed the activation of the pulmonary epithelium in response to B. anthracis spore exposure. Thus, the lung epithelia actively participate in the innate immune response to B. anthracis infection through cell signal-mediated elaboration of cytokines and chemokines.


Subject(s)
Anthrax/immunology , Bacillus anthracis/immunology , Immunity, Innate , Lung/immunology , Anthracenes/pharmacology , Butadienes/pharmacology , Chemokines/biosynthesis , Chemokines/immunology , Cytokines/biosynthesis , Cytokines/immunology , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/immunology , Gene Expression Regulation , Humans , Imidazoles/pharmacology , Immunohistochemistry , In Vitro Techniques , Lung/cytology , Lung/microbiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Macrophages, Alveolar/immunology , Microscopy, Fluorescence , Nitriles/pharmacology , Pulmonary Alveoli/pathology , Pyridines/pharmacology , RNA, Messenger/analysis , Spores, Bacterial/immunology
12.
Infect Immun ; 74(8): 4430-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16861629

ABSTRACT

Contact with the human alveolar macrophage plays a key role in the innate immune response to Bacillus anthracis spores. Because there is a significant delay between the initial contact of the spore with the host and clinical evidence of disease, there appears to be temporary containment of the pathogen by the innate immune system. Therefore, the early macrophage response to Bacillus anthracis exposure is important in understanding the pathogenesis of this disease. In this paper, we studied the initial events after exposure to spores, beginning with the rapid internalization of spores by the macrophages. Spore exposure rapidly activated the mitogen-activated protein kinase signaling pathways extracellular signal-regulated kinase, c-Jun-NH2-terminal kinase, and p38. This was followed by the transcriptional activation of cytokine and primarily monocyte chemokine genes as determined by RNase protection assays. Transcriptional induction is reflected at the translational level, as interleukin-1alpha (IL-1alpha), IL-1beta, IL-6, and tumor necrosis factor alpha (TNF-alpha) cytokine protein levels were markedly elevated as determined by enzyme-linked immunosorbent assay. Induction of IL-6 and TNF-alpha, and, to a lesser extent, IL-1alpha and IL-1beta, was partially inhibited by the blockade of individual mitogen-activated protein kinases, while the complete inhibition of cytokine induction was achieved when multiple signaling pathway inhibitors were used. Taken together, these data clearly show activation of the innate immune system in human alveolar macrophages by Bacillus anthracis spores. The data also show that multiple signaling pathways are involved in this cytokine response. This report is the first comprehensive examination of this process in primary human alveolar macrophages.


Subject(s)
Bacillus anthracis/immunology , Bacillus anthracis/physiology , Cytokines/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Mitogen-Activated Protein Kinases/metabolism , Chemokines/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunity, Innate , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , Phagocytosis , Spores, Bacterial/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...