Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
FASEB J ; 35(6): e21551, 2021 06.
Article in English | MEDLINE | ID: mdl-34042222

ABSTRACT

Intestinal epithelial cells (IEC) are crucial for maintaining proper digestion and overall homeostasis of the gut mucosa. IEC proliferation and differentiation are tightly regulated by well described pathways, however, relatively little is known about how cytokines shape these processes. Given that the anti-inflammatory cytokine interleukin (IL)-10 promotes intestinal barrier function, and insufficient IL-10 signaling increases susceptibility to intestinal diseases like inflammatory bowel disease, we hypothesized that IL-10 signaling modulates processes underlying IEC proliferation and differentiation. This was tested using in vivo and in vitro IEC-specific IL-10 receptor 1 (IL-10R1) depletion under homeostatic conditions. Our findings revealed that loss of IL-10R1 drove lineage commitment toward a dominant goblet cell phenotype while decreasing absorptive cell-related features. Diminished IL-10 signaling also significantly elevated IEC proliferation with relatively minor changes to apoptosis. Characterization of signaling pathways upstream of proliferation demonstrated a significant reduction in the Wnt inhibitor, DKK1, increased nuclear localization of ß-catenin, and increased transcripts of the proliferation marker, OLFM4, with IL-10R1 depletion. Phosphorylated STAT3 was nearly completely absent in IL-10R1 knockdown cells and may provide a mechanistic link between our observations and the regulation of these cellular processes. Our results demonstrate a novel role for IL-10 signaling in intestinal mucosal homeostasis by regulating proper balance of proliferation and IEC lineage fate.


Subject(s)
Cell Differentiation , Cell Proliferation , Epithelial Cells/pathology , Goblet Cells/pathology , Intestinal Mucosa/pathology , Receptors, Interleukin-10/physiology , Animals , Apoptosis , Epithelial Cells/metabolism , Female , Goblet Cells/metabolism , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
2.
Methods Mol Biol ; 2087: 79-91, 2020.
Article in English | MEDLINE | ID: mdl-31728984

ABSTRACT

Transmigration of neutrophils through an epithelial layer, such as in the intestine or lung, is a necessary response to a perceived attack at the mucosal surface of that tissue. This process is dynamically regulated by a number of interactive events between the neutrophil and other cell types and allows for an effective and localized neutrophil response. However, in certain inflammatory diseases, including inflammatory bowel disease and chronic obstructive pulmonary disease (COPD), persistent neutrophil accumulation can contribute to disease pathology. Elucidating the mechanisms of this aberrant neutrophil accumulation is crucial for understanding and ameliorating these disease processes. The method we describe here is a controlled model system that allows for the investigation of the interactive signals involved in neutrophil transmigration through epithelial barriers, and possible mechanisms of deregulation of this process.


Subject(s)
Epithelium/immunology , Epithelium/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Transendothelial and Transepithelial Migration/immunology , Cell Line , Cell Movement , Cell Separation , Cells, Cultured , Epithelial Cells , Humans
3.
Am J Physiol Gastrointest Liver Physiol ; 317(4): G531-G544, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31393789

ABSTRACT

Restitution of wounds in colonic epithelium is essential in the maintenance of health. Microbial products, such as the short-chain fatty acid butyrate, can have positive effects on wound healing. We used an in vitro model of T84 colonic epithelial cells to determine if the Snail genes Slug (SNAI2) and Snail (SNAI1), implemented in keratinocyte monolayer healing, are involved in butyrate-enhanced colonic epithelial wound healing. Using shRNA-mediated Slug/Snail knockdown, we found that knockdown of Slug (Slug-KD), but not Snail (Snail-KD), impairs wound healing in scratch assays with and without butyrate. Slug and Snail had differential effects on T84 monolayer barrier integrity, measured by transepithelial resistance, as Snail-KD impaired the barrier (with or without butyrate), whereas Slug-KD enhanced the barrier, again with or without butyrate. Targeted transcriptional analysis demonstrated differential expression of several tight junction genes, as well as focal adhesion genes. This included altered regulation of Annexin A2 and ITGB1 in Slug-KD, which was reflected in confocal microscopy, showing increased accumulation of B1-integrin protein in Slug-KD cells, which was previously shown to impair wound healing. Transcriptional analysis also indicated altered expression of genes associated with epithelial terminal differentiation, such that Slug-KD cells skewed toward overexpression of secretory cell pathway-associated genes. This included trefoil factors TFF1 and TFF3, which were expressed at lower than control levels in Snail-KD cells. Since TFFs can enhance the barrier in epithelial cells, this points to a potential mechanism of differential modulation by Snail genes. Although Snail genes are crucial in epithelial wound restitution, butyrate responses are mediated by other pathways as well.NEW & NOTEWORTHY Although butyrate can promote colonic mucosal healing, not all of its downstream pathways are understood. We show that the Snail genes Snail and Slug are mediators of butyrate responses. Furthermore, these genes, and Slug in particular, are necessary for efficient restitution of wounds and barriers in T84 epithelial cells even in the absence of butyrate. These effects are achieved in part through effects on regulation of ß1 integrin and cellular differentiation state.


Subject(s)
Butyrates/therapeutic use , Colonic Diseases/drug therapy , Colonic Diseases/genetics , Snail Family Transcription Factors/genetics , Wound Healing/drug effects , Wound Healing/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line , Gene Knockdown Techniques , Humans , Signal Transduction/drug effects , Tight Junction Proteins/drug effects , Tight Junction Proteins/genetics , Trefoil Factor-1/biosynthesis , Trefoil Factor-1/genetics , Trefoil Factor-3/biosynthesis , Trefoil Factor-3/genetics
4.
Am J Pathol ; 186(2): 259-69, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26687815

ABSTRACT

Pulmonary hypertension subsequent to an infectious disease can be due to vascular structural remodeling or to functional alterations within various vascular cell types. In our previous mouse model of Pneumocystis-associated pulmonary hypertension, we found that vascular remodeling was not responsible for observed increases in right ventricular pressures. Here, we report that the vascular dysfunction we observed could be explained by an enhanced response to endothelin-1 (20% greater reduction in lumen diameter, P ≤ 0.05), corresponding to an up-regulation of similar magnitude (P ≤ 0.05) of the endothelin A receptor in the lung tissue. This effect was potentially augmented by a decrease in production of the pulmonary vasodilator adrenomedullin of almost 70% (P ≤ 0.05). These changes did not occur in interferon-γ knockout mice similarly treated, which do not develop pulmonary hypertension under these circumstances. Surprisingly, we did not observe any relevant changes in the vascular endothelial nitric oxide synthase vasodilatory response, which is a common potential site of inflammatory alterations to pulmonary vascular function. Our results indicate the diverse mechanisms by which inflammatory responses to prior infections can cause functionally relevant changes in vascular responses in the lung, promoting the development of pulmonary hypertension.


Subject(s)
Adrenomedullin/metabolism , Endothelins/metabolism , Hypertension, Pulmonary/metabolism , Lung/metabolism , Pneumocystis/metabolism , Pulmonary Artery/metabolism , Adrenomedullin/genetics , Animals , Hypertension, Pulmonary/physiopathology , Interferon-gamma/metabolism , Mice, Inbred BALB C , Nitric Oxide Synthase Type III/metabolism , Pulmonary Artery/physiopathology , Up-Regulation , Vasodilator Agents/pharmacology
5.
Am J Pathol ; 184(2): 483-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24361497

ABSTRACT

Pulmonary hypertension (PH) is a disease of diverse etiology. Although primary PH can develop in the absence of prior disease, PH more commonly develops in conjunction with other pulmonary pathologies. We previously reported a mouse model in which PH occurs as a sequela of Pneumocystis infection in the context of transient CD4 depletion. Here, we report that instead of the expected Th2 pathways, the Th1 cytokine IFN-γ is essential for the development of PH, as wild-type mice developed PH but IFN-γ knockout mice did not. Because gene expression analysis showed few strain differences that were not immune-function related, we focused on those responses as potential pathologic mechanisms. In addition to dependence on IFN-γ, we found that when CD4 cells were continuously depleted, but infection was limited by antibiotic treatment, PH did not occur, confirming that CD4 T cells are required for PH development. Also, although CD8 T-cells are implicated in the pathology of Pneumocystis pneumonia, they did not have a role in the onset of PH. Finally, we found differences in immune cell phenotypes that correlated with PH, including elevated CD204 expression in lung CD11c(+) cells, but their role remains unclear. Overall, we demonstrate that a transient, localized, immune response requiring IFN-γ and CD4-T cells can disrupt pulmonary vascular function and promote lingering PH.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/microbiology , Interferon-gamma/metabolism , Pneumocystis/physiology , Animals , Bronchoalveolar Lavage Fluid , Fibrosis , Gene Expression Regulation , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/pathology , Immunity/genetics , Immunophenotyping , Interleukin-12/metabolism , Lymphocyte Depletion , Mice , Mice, Knockout , Pneumonia, Pneumocystis/complications , Pneumonia, Pneumocystis/genetics , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/pathology , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/metabolism , Scavenger Receptors, Class A/metabolism , Up-Regulation
6.
Am J Respir Cell Mol Biol ; 46(3): 290-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21960549

ABSTRACT

It is widely held that exposure to pathogens such as fungi can be an agent of comorbidity, such as exacerbation of asthma or chronic obstructive pulmonary disease. Although many studies have examined allergic responses to fungi and their effects on pulmonary function, the possible pathologic implications of the early innate responses to fungal pathogens have not been explored. We examined early responses to the atypical fungus Pneumocystis in two common strains of mice in terms of overall immunological response and related pathology, such as cell damage and airway hyperresponsiveness (AHR). We found a strong strain-specific response in BALB/c mice that included recruitment of neutrophils, NK, NKT, and CD4 T cells. This response was accompanied by elevated indicators of lung damage (bronchoalveolar lavage fluid albumin and LDH) and profound AHR. This early response was absent in C57BL/6 mice, although both strains exhibited a later response associated with the clearance of Pneumocystis. We found that this AHR could not be attributed exclusively to the presence of recruited neutrophils, NKT, NK, or CD4 cells or to the actions of IFN-γ or IL-4. However, in the absence of STAT6 signaling, AHR and inflammatory cell recruitment were virtually absent. Gene expression analysis indicated that this early response included activation of several transcription factors that could be involved in pulmonary remodeling. These results show that exposure to a fungus such as Pneumocystis can elicit pulmonary responses that may contribute to morbidity, even without prior sensitization, in the context of certain genetic backgrounds.


Subject(s)
Bronchial Hyperreactivity/metabolism , Immunity, Innate , Lung Diseases, Fungal/metabolism , Lung/metabolism , Pneumocystis Infections/metabolism , STAT6 Transcription Factor/metabolism , Albumins/metabolism , Animals , Antigens, CD1/genetics , Antigens, CD1/metabolism , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/microbiology , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/microbiology , Disease Models, Animal , Gene Expression Regulation , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-4/metabolism , L-Lactate Dehydrogenase/metabolism , Lung/immunology , Lung/microbiology , Lung/physiopathology , Lung Diseases, Fungal/genetics , Lung Diseases, Fungal/immunology , Lung Diseases, Fungal/microbiology , Lung Diseases, Fungal/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/microbiology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Pneumocystis Infections/genetics , Pneumocystis Infections/immunology , Pneumocystis Infections/microbiology , Pneumocystis Infections/physiopathology , Receptors, Interleukin-4/deficiency , Receptors, Interleukin-4/genetics , Receptors, Interleukin-8B/deficiency , Receptors, Interleukin-8B/genetics , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics , Signal Transduction , Species Specificity , Time Factors
7.
Infect Immun ; 79(5): 1905-14, 2011 May.
Article in English | MEDLINE | ID: mdl-21343358

ABSTRACT

Infection with the opportunistic fungal pathogen Pneumocystis is assumed to pass without persistent pathology in immunocompetent hosts. However, when immunocompetent BALB/c mice were inoculated with Pneumocystis, a vigorous Th2-like pulmonary inflammation ensued and peaked at 14 days postinfection. This coincided with a 10-fold increase in the number of antigen-presenting cells (APCs) in the lung, and these cells were capable of presenting antigen in vitro, as well as greater uptake of antigen in vivo. When mice were presented with exogenous antigen at the 14-day time point of the infection, they developed respiratory sensitization to that antigen, in the form of increased airway hyperresponsiveness upon a later challenge, whereas mice not infected but presented with antigen did not. Like other forms of collateral sensitization, this response was dependent on interleukin-4 receptor signaling. This ability to facilitate sensitization to exogenous antigen has been previously reported for other infectious disease agents; however, Pneumocystis appears to be uniquely capable in this respect, as a single intranasal dose without added adjuvant, when it was administered at the appropriate time, was sufficient to initiate sensitization. Pneumocystis infection probably occurs in most humans during the first few years of life, and in the vast majority of cases, it fails to cause any overt direct pathology. However, as we show here, Pneumocystis can be an agent of comorbidity at this time by facilitating respiratory sensitization that may relate to the later development or exacerbation of obstructive airway disease.


Subject(s)
Antigens/immunology , Pneumocystis Infections/immunology , Respiratory Hypersensitivity/immunology , Animals , Cell Separation , Cytokines/immunology , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/immunology , Receptors, Interleukin-4/deficiency , Receptors, Interleukin-4/immunology
8.
PLoS One ; 4(9): e7142, 2009 Sep 23.
Article in English | MEDLINE | ID: mdl-19774076

ABSTRACT

BACKGROUND: Destruction of the architectural and subsequently the functional integrity of the lung following pulmonary viral infections is attributable to both the extent of pathogen replication and to the host-generated inflammation associated with the recruitment of immune responses. The presence of antigenically disparate pulmonary viruses and the emergence of novel viruses assures the recurrence of lung damage with infection and resolution of each primary viral infection. Thus, there is a need to develop safe broad spectrum immunoprophylactic strategies capable of enhancing protective immune responses in the lung but which limits immune-mediated lung damage. The immunoprophylactic strategy described here utilizes a protein cage nanoparticle (PCN) to significantly accelerate clearance of diverse respiratory viruses after primary infection and also results in a host immune response that causes less lung damage. METHODOLOGY/PRINCIPAL FINDINGS: Mice pre-treated with PCN, independent of any specific viral antigens, were protected against both sub-lethal and lethal doses of two different influenza viruses, a mouse-adapted SARS-coronavirus, or mouse pneumovirus. Treatment with PCN significantly increased survival and was marked by enhanced viral clearance, accelerated induction of viral-specific antibody production, and significant decreases in morbidity and lung damage. The enhanced protection appears to be dependent upon the prior development of inducible bronchus-associated lymphoid tissue (iBALT) in the lung in response to the PCN treatment and to be mediated through CD4+ T cell and B cell dependent mechanisms. CONCLUSIONS/SIGNIFICANCE: The immunoprophylactic strategy described utilizes an infection-independent induction of naturally occurring iBALT prior to infection by a pulmonary viral pathogen. This strategy non-specifically enhances primary immunity to respiratory viruses and is not restricted by the antigen specificities inherent in typical vaccination strategies. PCN treatment is asymptomatic in its application and importantly, ameliorates the damaging inflammation normally associated with the recruitment of immune responses into the lung.


Subject(s)
Bronchi/microbiology , Bronchi/virology , Lung/virology , Lymphoid Tissue/metabolism , Nanoparticles/chemistry , Nanotechnology/methods , Orthomyxoviridae/metabolism , Pneumovirus/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Animals , Bronchi/metabolism , Bronchoalveolar Lavage Fluid , Coxiella burnetii/metabolism , Female , Infections/pathology , Lung/immunology , Lung/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
9.
Am J Pathol ; 171(3): 790-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17640969

ABSTRACT

Improved treatment regimens have reduced fatalities from opportunistic diseases, such as Pneumocystis pneumonia, in AIDS patients. However, serious chronic conditions, including pulmonary hypertension (PH), are increasing in this group. We report here that when CD4 T cells in Pneumocystis-infected mice are temporally depleted and then allowed to return, the extended inflammation results in PH that persists after Pneumocystis is eliminated. Using this model of PH, we have found that i) the onset of PH is correlated with the return of CD4 T cells, but PH persists after CD4 levels diminish; ii) vascular remodeling accompanies PH, but whereas temporary medial hypertrophy is evident with transient PH in immunocompetent mice, persistent PH is associated with perivascular fibrosis; iii) elevated levels of the fibrotic mediator FIZZ1 are found in bronchoalveolar lavage fluid of mice with persistent PH; and iv) although Th2-related mechanisms may be involved in PH etiology, PH still occurs in interleukin-4 receptor-deficient mice under these conditions. Overall, the data presented here demonstrate that the immune response to an infectious disease pathogen, such as Pneumocystis, can, when perturbed and prolonged, lead to later development of a serious chronic condition such as PH.


Subject(s)
Hypertension, Pulmonary/etiology , Pneumonia, Pneumocystis/complications , Animals , Bronchoalveolar Lavage Fluid/cytology , CD4-Positive T-Lymphocytes/metabolism , Fibrosis/pathology , HIV Infections/complications , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Inflammation/metabolism , Lung/cytology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/pathology , Pulmonary Artery/cytology , Pulmonary Artery/pathology , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , Signal Transduction/physiology
10.
Am J Pathol ; 168(2): 466-75, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16436661

ABSTRACT

Pneumocystis spp. pneumonia (PCP) in humans and in surrogate animal species typically occurs in the absence of CD4 T cells, as takes place during acquired immune deficiency syndrome. However, patients treated with highly active anti-retroviral therapy sometimes exhibit an exacerbation of diseases such as PCP that coincides with resurgent CD4 T cells, a phenomenon known as immune reconstitution disease. We used an animal model of PCP using the B-cell-deficient muMT mouse together with antibody-mediated depletion of various T-cell subsets to examine the role of CD4 and CD8 T cells in the development of pathology in PCP. Although overt pathology occurs in the presence of CD4 T cells only, CD8 T cells only, or both, pulmonary injury occurs via different paths, depending on the complement of T cells present. Surprisingly, profound damage occurred when only CD4 T cells were present, and this pathology coincided with enhanced recruitment and activation of eosinophils and strong type 2 cytokine polarization in the alveolar environment. In addition, CD8 T cells can act to moderate this CD4 T cell-mediated pathology, possibly by increasing the ratio of putative CD25+ suppressor CD4 T cells to CD25- effector CD4 T cells.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Eosinophils/immunology , Pneumonia, Pneumocystis/pathology , Animals , B-Lymphocytes/pathology , Bronchoalveolar Lavage , Flow Cytometry , Lung/pathology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/immunology , Pulmonary Alveoli/microbiology , Pulmonary Alveoli/pathology
11.
Infect Immun ; 72(10): 5722-32, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15385471

ABSTRACT

Neutrophils are implicated in the damage of lung tissue in many disease states, including infectious diseases and environmental insults. These effects may be due to oxidative or nonoxidative functions of the neutrophil or both. We examined the role of neutrophils in pulmonary damage during infection with the opportunistic fungal pathogen Pneumocystis sp. in four mouse models of neutrophil dysfunction. These were (i) a knockout of the gp91(phox) component of NADPH oxidase, in which reactive oxygen species (ROS) production is greatly reduced; (ii) a double knockout of gp91(phox) and inducible nitric oxide synthase, in which ROS and nitric oxide production is greatly decreased; (iii) a knockout of the chemokine receptor CXCR2, in which accumulation of intra-alveolar neutrophils is severely diminished; and (iv) antibody depletion of circulating neutrophils in wild-type mice with the monoclonal antibody RB6. Surprisingly, in each case, indicators of pulmonary damage (respiratory rates, arterial oxygen partial pressures, and intra-alveolar albumin concentrations) were the same in knockout mice and comparable wild-type mice. Therefore, whereas neutrophils are a valid correlative marker of lung damage during Pneumocystis infection, neither neutrophils nor ROS appear to be the causative agent of tissue damage. We also show that there is no difference in Pneumocystis burdens between wild-type and knockout mice, which supports the idea that neutrophils do not have a major role in the clearance of this organism.


Subject(s)
Neutrophils/physiology , Pneumonia, Pneumocystis/metabolism , Pneumonia, Pneumocystis/pathology , Reactive Oxygen Species/metabolism , Animals , Bronchoalveolar Lavage , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Gene Deletion , Inflammation/immunology , Inflammation/pathology , Inflammation/physiopathology , Leukocyte Count , Lung/immunology , Lung/pathology , Lung/physiopathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Neutrophils/cytology , Neutrophils/immunology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/physiopathology , Receptors, Interleukin-8B/deficiency , Receptors, Interleukin-8B/genetics , Respiration
12.
Infect Immun ; 71(11): 6213-21, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14573639

ABSTRACT

Host defense against the opportunistic pathogen Pneumocystis carinii requires functional interactions of many cell types. Alveolar macrophages are presumed to be a vital host cell in the clearance of P. carinii, and the mechanisms of this interaction have come under scrutiny. The macrophage mannose receptor is believed to play an important role as a receptor involved in the binding and phagocytosis of P. carinii. Although there is in vitro evidence for this interaction, the in vivo role of this receptor in P. carinii clearance in unclear. Using a mouse model in which the mannose receptor has been deleted, we found that the absence of this receptor is not sufficient to allow infection by P. carinii in otherwise immunocompetent mice. Furthermore, when mice were rendered susceptible to P. carinii by CD4(+) depletion, mannose receptor knockout mice (MR-KO) had pathogen loads equal to those of wild-type mice. However, the MR-KO mice exhibited a greater influx of phagocytes into the alveoli during infection. This was accompanied by increased pulmonary pathology in the MR-KO mice, as well as greater accumulation of glycoproteins in the alveoli (glycoproteins, including harmful hydrolytic enzymes, are normally cleared by the mannose receptor). We also found that the surface expression of the mannose receptor is not downregulated during P. carinii infection in wild-type mice. Our findings suggest that while the macrophage mannose receptor may be important in the recognition of P. carinii, in vivo, this mechanism may be redundant, and the absence of this receptor may be compensated for.


Subject(s)
Lectins, C-Type/physiology , Macrophages/physiology , Mannose-Binding Lectins/physiology , Pneumocystis Infections/immunology , Receptors, Cell Surface/physiology , Animals , CD4-Positive T-Lymphocytes/physiology , Disease Susceptibility , Mannose/metabolism , Mannose Receptor , Mice , Mice, Inbred C57BL , Mice, Knockout , Serum Albumin, Bovine/metabolism
13.
Inflammation ; 27(1): 45-58, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12772776

ABSTRACT

Adenosine is a potent inhibitor of reactive oxygen species (ROS) production by the NADPH oxidase in fMLF-stimulated neutrophils. Although much is known about the pharamacology and signal transduction of this effect, it is not known how adenosine affects assembly and localization of the NADPH oxidase components within the neutrophil. We report here that adenosine pretreatment of fMLF-stimulated neutrophils results in decreased plasma membrane/secretory granule content of the flavocytochrome b components (p22phox and gp91phox) of the NADPH oxidase, which correlates with inhibition of ROS production. Adenosine treatment did not affect upregulation of secretory and specific granule surface markers, confirming that degranulation was not impaired by adenosine. However, adenosine treatment did result in increased movement of cell-surface flavocytochrome b to heavy granule fractions in fMLF-stimulated neutrophils. These data suggest that adenosine-mediated effects on neutrophil ROS production are due, in part to endocytosis and/or redistribution of flavocytochrome b between various subcellular compartments.


Subject(s)
Adenosine/pharmacology , Cytochromes b/metabolism , Membrane Transport Proteins , NADPH Oxidases/antagonists & inhibitors , Neutrophils/enzymology , Subcellular Fractions/metabolism , Cell Membrane/metabolism , Cytochromes b/analysis , Humans , Membrane Glycoproteins/analysis , Membrane Glycoproteins/metabolism , N-Formylmethionine Leucyl-Phenylalanine , NADPH Dehydrogenase/analysis , NADPH Dehydrogenase/metabolism , NADPH Oxidase 2 , Neutrophil Activation , Neutrophils/metabolism , Neutrophils/ultrastructure , Phosphoproteins/analysis , Phosphoproteins/metabolism , Protein Transport , Reactive Oxygen Species/metabolism , Secretory Vesicles/metabolism
14.
J Leukoc Biol ; 73(3): 344-55, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12629148

ABSTRACT

Peroxynitrite, a potent oxidant generated in inflammatory tissues, can nitrate tyrosine residues on a variety of proteins. Based on previous studies suggesting that actin might be a potential target for peroxynitrite-mediated nitration in neutrophils, we investigated the effects of peroxynitrite on actin function. We show here that peroxynitrite and the peroxynitrite generator (SIN-1) modified actin in a concentration-dependent manner, resulting in an inhibition of globular-actin polymerization and filamentous-actin depolymerization in vitro. The effects of peroxynitrite were inhibited by the pyrrolopyrimidine antioxidant PNU-101033E, which has been shown previously to specifically block peroxynitrite-mediated tyrosine nitration. Furthermore, spectrophotometric and immunoblot analysis of peroxynitrite-treated actin demonstrated a concentration-dependent increase in nitrotyrosine, which was also blocked by PNU-101033E. Activation of neutrophils in the presence of a nitric oxide donor (S-nitroso-N-acetylpenicillamine) resulted in nitration of exogenously added actin. Nitrated actin was also found in peroxynitrite-treated neutrophils, suggesting that actin may be an important intracellular target during inflammation. To investigate this issue, we analyzed the effect of peroxynitrite treatment on a number of actin-dependent neutrophil processes. Indeed, neutrophil actin polymerization, migration, phagocytosis, and respiratory burst activity were all inhibited by SIN-1 treatment in a concentration-dependent manner. Therefore, the ability of peroxynitrite to inhibit actin dynamics has a significant effect on actin-dependent, cellular processes in phagocytic cells and may modulate their host defense function.


Subject(s)
Actins/antagonists & inhibitors , Molsidomine/analogs & derivatives , Neutrophils/drug effects , Peroxynitrous Acid/pharmacology , Tyrosine/analogs & derivatives , Actins/metabolism , Chemotaxis/drug effects , Dimerization , Dose-Response Relationship, Drug , Humans , Molsidomine/pharmacology , Neutrophil Activation , Neutrophils/physiology , Neutrophils/ultrastructure , Nitric Oxide Donors , Phagocytosis/drug effects , Reactive Oxygen Species/metabolism , Respiratory Burst/drug effects , Tyrosine/analysis
15.
Article in English | MEDLINE | ID: mdl-12223206

ABSTRACT

During the host defense process, neutrophils migrate into infected tissues where they become activated, resulting in the assembly of a superoxide anion-generating complex known as the NADPH oxidase. Despite the importance of this system in animal host defense, almost nothing is known about the NADPH oxidase in neutrophils from wild ruminant species. In the present studies, we provide a molecular analysis of the bison leukocyte NADPH oxidase. Using reverse transcriptase-polymerase chain reaction and rapid amplification of cDNA ends, we cloned and sequenced the full-length cDNAs for five bison NADPH oxidase components: p22(phox), p40(phox), p47(phox) and p67(phox), and gp91(phox). When compared to other species, the deduced amino acid sequences of the bison homologs were most similar to those of bovine. Interestingly, a bison p40(phox) alternative splice product was isolated, which was similar to that observed for human p40(phox) in that the cDNAs contained sequence from intron 8. Consistent with the high degree of similarity between bison and bovine amino acid sequences, immunoblot analysis showed that the bison homologs migrated similarly to their bovine counterparts. Overall, these studies show that the bison and bovine NADPH oxidase genes are highly conserved between these two species, despite their divergence from a common ancestor over 1 million years ago.


Subject(s)
Bison/genetics , DNA, Complementary/chemistry , Membrane Transport Proteins , NADPH Oxidases/genetics , Phagocytes/enzymology , Alternative Splicing , Amino Acid Sequence , Animals , Cloning, Molecular , Membrane Glycoproteins/genetics , Molecular Sequence Data , NADPH Dehydrogenase/genetics , NADPH Oxidase 2 , NADPH Oxidases/biosynthesis , Neutrophils/enzymology , Phosphoproteins/genetics , Sequence Alignment , Sequence Analysis, DNA
16.
J Leukoc Biol ; 71(5): 764-74, 2002 May.
Article in English | MEDLINE | ID: mdl-11994500

ABSTRACT

The neutrophil inflammatory response can be altered profoundly by contact with extracellular matrix proteins (ECMs). We characterized functional responses (intracellular calcium, actin polymerization, degranulation, adhesion, and oxidative burst) of bovine neutrophils adhered to selected ECM proteins [collagen IV, laminin, fibronectin, thrombospondin, and heparan sulfate proteoglycan (HSP)] in response to interleukin-8 (IL-8) and platelet-activating factor (PAF). Neutrophil adhesion to ECMs altered responses to PAF and IL-8, although some functions were more responsive to modulation. The most susceptible function was reactive oxygen species (ROS) production. ROS production in response to PMA and TNF-alpha was supported differentially by various ECMs, and PAF and IL-8 "priming" had strikingly different effects, depending on the ECM present. Although PAF and IL-8 inhibited TNF-alpha-induced ROS production in neutrophils adhered to collagen, fibronectin, and laminin, PAF enhanced ROS production strongly in HSP-adherent cells. This study illustrates how neutrophils can integrate multiple stimuli, resulting in complex modulation of their functional response.


Subject(s)
Cell Adhesion , Extracellular Matrix Proteins/pharmacology , Inflammation Mediators/pharmacology , Neutrophil Activation , Neutrophils/immunology , Actin Cytoskeleton/ultrastructure , Animals , Calcium/metabolism , Cattle , Cell Degranulation , Cells, Cultured , Extracellular Matrix Proteins/metabolism , Interleukin-8/pharmacology , Kinetics , Neutrophils/drug effects , Neutrophils/ultrastructure , Platelet Activating Factor/pharmacology , Reactive Oxygen Species/metabolism
17.
Antioxid Redox Signal ; 4(1): 69-83, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11970845

ABSTRACT

Neutrophils play an essential role in the body's innate immune response to infection. To protect the host, these phagocytic cells possess an impressive array of microbicidal weapons that can be brought to bear on an invading pathogen, including a variety of toxic oxygen radical species and proteolytic enzymes. Although the neutrophil response is designed to restrict the damage to the smallest possible region where the pathogen is located, some of the damaging agents inevitably leak into the surrounding areas where they have the capacity to inflict tissue damage at sites of inflammation. Thus, it is essential that the host defense response of these cells is finely tuned to result in the appropriate level of response to any given situation. One of the regulatory mechanisms implicated in controlling neutrophil responses is priming. Through the action of priming agents, the level of activation and subsequent responses of the cell can be regulated so that a continuum of activation states is achieved. In this review, we describe key features of the priming response in host defense and disease pathogenesis and focus on the unique role of reactive oxygen species as priming agents.


Subject(s)
Neutrophil Activation , Neutrophils/immunology , Neutrophils/physiology , Oxygen/metabolism , Animals , Enzyme Activation , Humans , Models, Biological , Reactive Oxygen Species
SELECTION OF CITATIONS
SEARCH DETAIL
...