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










Publication year range
1.
Infect Immun ; 88(2)2020 01 22.
Article in English | MEDLINE | ID: mdl-31611280

ABSTRACT

The pulmonary immune response protects healthy individuals against Pneumocystis pneumonia (PcP). However, the immune response also drives immunopathogenesis in patients who develop severe PcP, and it is generally accepted that optimal treatment requires combination strategies that promote fungal killing and also provide effective immunomodulation. The anti-inflammatory drug sulfasalazine programs macrophages for enhanced Pneumocystis phagocytosis and also suppresses PcP-related immunopathogenesis. Anti-Pneumocystis antibody opsonizes Pneumocystis organisms for greater phagocytosis and may also mask antigens that drive immunopathogenesis. Thus, we hypothesized that combining antibody and sulfasalazine would have the dual benefit of enhancing fungal clearance while dampening immunopathogenesis and allow the rescue of severe PcP. To model a clinically relevant treatment scenario in mice, therapeutic interventions were withheld until clear symptoms of pneumonia were evident. When administered individually, both passive antibody and sulfasalazine improved pulmonary function and enhanced Pneumocystis clearance to similar degrees. However, combination treatment with antibody and sulfasalazine produced a more rapid improvement, with recovery of body weight, a dramatic improvement in pulmonary function, reduced lung inflammation, and the rapid clearance of the Pneumocystis organisms. Accelerated fungal clearance in the combination treatment group was associated with a significant increase in macrophage phagocytosis of Pneumocystis Both passive antibody and sulfasalazine resulted in the suppression of Th1 cytokines and a marked increase in lung macrophages displaying an alternatively activated phenotype, which were enhanced by combination treatment. Our data support the concept that passive antibody and sulfasalazine could be an effective and specific adjunctive therapy for PcP, with the potential to accelerate fungal clearance while attenuating PcP-associated immunopathogenesis.


Subject(s)
Antibodies/immunology , Fungi/drug effects , Fungi/immunology , Pneumonia, Pneumocystis/drug therapy , Pneumonia, Pneumocystis/immunology , Sulfasalazine/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cytokines/immunology , Female , Immunologic Factors/immunology , Immunomodulation/drug effects , Immunomodulation/immunology , Immunotherapy/methods , Inflammation/immunology , Lung/drug effects , Lung/immunology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Mice , Mice, SCID , Phagocytosis/drug effects , Phagocytosis/immunology
2.
Front Immunol ; 9: 2131, 2018.
Article in English | MEDLINE | ID: mdl-30283457

ABSTRACT

Invasive fungal infections, including Pneumocystis Pneumonia (PcP), remain frequent life-threatening conditions of patients with adaptive immune defects. While innate immunity helps control pathogen growth early during infection, it is typically not sufficient for complete protection against Pneumocystis and other human fungal pathogens. Alveolar macrophages (AM) possess pattern recognition molecules capable of recognizing antigenic and structural determinants of Pneumocystis. However, this pathogen effectively evades innate immunity to infect both immunocompetent and immunosuppressed hosts, albeit with differing outcomes. During our studies of mouse models of PcP, the FVB/N strain was identified as unique because of its ability to mount a protective innate immune response against Pneumocystis infection. In contrast to other immunocompetent strains, which become transiently infected prior to the onset of adaptive immunity, FVB/N mice rapidly eradicated Pneumocystis before an adaptive immune response was triggered. Furthermore, FVB/N mice remained highly resistant to infection even in the absence of functional T cells. The effector mechanism of innate protection required the action of functional alveolar macrophages, and the adoptive transfer of resistant FVB/N AMs, but not susceptible CB.17 AMs, conferred protection to immunodeficient mice. Macrophage IFNγ receptor signaling was not required for innate resistance, and FVB/N macrophages were found to display markers of alternative activation. IFNγ reprogrammed resistant FVB/N macrophages to a permissive M1 biased phenotype through a mechanism that required direct activation of the macrophage IFNγR. These results demonstrate that appropriately programmed macrophages provide protective innate immunity against this opportunistic fungal pathogen, and suggest that modulating macrophage function may represent a feasible therapeutic strategy to enhance antifungal host defense. The identification of resistant and susceptible macrophages provides a novel platform to study not only the mechanisms of macrophage-mediated antifungal defense, but also the mechanisms by which Pneumocystis evades innate immunity.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Macrophages, Alveolar/immunology , Pneumocystis Infections/immunology , Animals , Humans , Immunocompetence , Immunocompromised Host , Macrophages/immunology , Macrophages/metabolism , Macrophages, Alveolar/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Pneumocystis Infections/microbiology , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
Australas Psychiatry ; 25(5): 494-496, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28836814

ABSTRACT

OBJECTIVE: The aim of this paper is to provide an overview of the delayed neurologic syndrome of carbon monoxide poisoning and its clinical importance in psychiatric settings. METHOD: A brief review of carbon monoxide poisoning is presented with a focus on the delayed neurologic syndrome and a case of deliberate self- poisoning is described. RESULTS: As in the case described, the delayed manifestations of carbon monoxide poisoning can resemble a relapse of psychiatric illness. CONCLUSIONS: In cases of carbon monoxide poisoning it is important to consider the delayed neurologic syndrome as misdiagnosis could lead to inappropriate treatment and worsened outcome.


Subject(s)
Carbon Monoxide Poisoning/diagnosis , Depressive Disorder, Major/diagnosis , Diagnostic Errors , Neurotoxicity Syndromes/diagnosis , Self-Injurious Behavior/diagnosis , Suicide, Attempted , Carbon Monoxide Poisoning/complications , Humans , Male , Middle Aged , Neurotoxicity Syndromes/etiology
4.
Cell Death Differ ; 24(3): 559-570, 2017 03.
Article in English | MEDLINE | ID: mdl-28060378

ABSTRACT

The phagocytosis of apoptotic cells (efferocytosis) shifts macrophages to an anti-inflammatory state through a set of still poorly understood soluble and cell-bound signals. Apoptosis is a common feature of inflamed tissues, and efferocytosis by tissue macrophages is thought to promote the resolution of inflammation. However, it is not clear how the exposure of tissue macrophages to inflammatory cues (e.g., PAMPs, DAMPs) in the early stages of inflammation affects immune outcomes of macrophage-apoptotic cell interactions occurring at later stages of inflammation. To address this, we used low-dose endotoxin conditioning (LEC, 1 ng/ml LPS 18 h) of mouse resident peritoneal macrophages (RPMФ) to model the effects of suboptimal (i.e., non-tolerizing), antecedent TLR activation on macrophage inflammatory responses to apoptotic cells. Compared with unconditioned macrophages (MФ), LEC-MФ showed a significant enhancement of apoptotic cell-driven suppression of many inflammatory cytokines (e.g., TNF, MIP-1ß, MCP-1). We then found that enzymatic depletion of adenosine or inhibition of the adenosine receptor A2a on LEC-MФ abrogated apoptotic cell suppression of TNF, and this suppression was entirely dependent on the ecto-enzyme CD73 (AMPadenosine) but not CD39 (ATPAMP), both of which are highly expressed on RPMФ. In addition to a requirement for CD73, we also show that Adora2a levels in macrophages are a critical determinant of TNF suppression by apoptotic cells. LEC treatment of RPMФ led to a ~3-fold increase in Adora2a and a ~28-fold increase in adenosine sensitivity. Moreover, in RAW264.7 cells, ectopic expression of both A2a and CD73 was required for TNF suppression by apoptotic cells. In mice, mild, TLR4-dependent inflammation in the lungs and peritoneum caused a rapid increase in macrophage Adora2a and Adora2b levels, and CD73 was required to limit neutrophil influx in this peritonitis model. Thus immune signaling via the CD73-A2a axis in macrophages links early inflammatory events to subsequent immune responses to apoptotic cells.


Subject(s)
5'-Nucleotidase/metabolism , Apoptosis/drug effects , Lipopolysaccharides/toxicity , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Adenosine/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apyrase/deficiency , Apyrase/genetics , Apyrase/metabolism , Cells, Cultured , Chemokine CXCL10/metabolism , Chemokine CXCL2/metabolism , Coculture Techniques , Cytokines/metabolism , Lung/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
5.
Infect Immun ; 83(12): 4594-603, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26371121

ABSTRACT

Pneumocystis is a respiratory fungal pathogen that causes pneumonia (Pneumocystis pneumonia [PcP]) in immunocompromised patients. Alveolar macrophages are critical effectors for CD4(+) T cell-dependent clearance of Pneumocystis, and previous studies found that alternative macrophage activation accelerates fungal clearance during PcP-related immune reconstitution inflammatory syndrome (IRIS). However, the requirement for either classically or alternatively activated macrophages for Pneumocystis clearance has not been determined. Therefore, RAG2(-/-) mice lacking either the interferon gamma (IFN-γ) receptor (IFN-γR) or interleukin 4 receptor alpha (IL-4Rα) were infected with Pneumocystis. These mice were then immune reconstituted with wild-type lymphocytes to preserve the normal T helper response while preventing downstream effects of Th1 or Th2 effector cytokines on macrophage polarization. As expected, RAG2(-/-) mice developed severe disease but effectively cleared Pneumocystis and resolved IRIS. Neither RAG/IFN-γR(-/-) nor RAG/IL-4Rα(-/-) mice displayed impaired Pneumocystis clearance. However, RAG/IFN-γR(-/-) mice developed a dysregulated immune response, with exacerbated IRIS and greater pulmonary function deficits than those in RAG2 and RAG/IL-4Rα(-/-) mice. RAG/IFN-γR(-/-) mice had elevated numbers of lung CD4(+) T cells, neutrophils, eosinophils, and NK cells but severely depressed numbers of lung CD8(+) T suppressor cells. Impaired lung CD8(+) T cell responses in RAG/IFN-γR(-/-) mice were associated with elevated lung IFN-γ levels, and neutralization of IFN-γ restored the CD8 response. These data demonstrate that restricting the ability of macrophages to polarize in response to Th1 or Th2 cytokines does not impair Pneumocystis clearance. However, a cell type-specific IFN-γ/IFN-γR-dependent mechanism regulates CD8(+) T suppressor cell recruitment, limits immunopathogenesis, preserves lung function, and enhances the resolution of PcP-related IRIS.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Reconstitution Inflammatory Syndrome/immunology , Macrophages, Alveolar/immunology , Pneumocystis/immunology , Pneumonia, Pneumocystis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Eosinophils/immunology , Eosinophils/microbiology , Eosinophils/pathology , Gene Expression Regulation , Host-Pathogen Interactions , Immune Reconstitution Inflammatory Syndrome/genetics , Immune Reconstitution Inflammatory Syndrome/microbiology , Immune Reconstitution Inflammatory Syndrome/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophage Activation , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Mice , Mice, Knockout , Mice, SCID , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Pneumocystis/pathogenicity , Pneumonia, Pneumocystis/genetics , Pneumonia, Pneumocystis/microbiology , Pneumonia, Pneumocystis/pathology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer/microbiology , T-Lymphocytes, Helper-Inducer/pathology , Th1-Th2 Balance , Interferon gamma Receptor
6.
PLoS Pathog ; 6(8): e1001058, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20808846

ABSTRACT

Although T cells are critical for host defense against respiratory fungal infections, they also contribute to the immunopathogenesis of Pneumocystis pneumonia (PcP). However, the precise downstream effector mechanisms by which T cells mediate these diverse processes are undefined. In the current study the effects of immune modulation with sulfasalazine were evaluated in a mouse model of PcP-related Immune Reconstitution Inflammatory Syndrome (PcP-IRIS). Recovery of T cell-mediated immunity in Pneumocystis-infected immunodeficient mice restored host defense, but also initiated the marked pulmonary inflammation and severe pulmonary function deficits characteristic of IRIS. Sulfasalazine produced a profound attenuation of IRIS, with the unexpected consequence of accelerated fungal clearance. To determine whether macrophage phagocytosis is an effector mechanism of T cell-mediated Pneumocystis clearance and whether sulfasalazine enhances clearance by altering alveolar macrophage phagocytic activity, a novel multispectral imaging flow cytometer-based method was developed to quantify the phagocytosis of Pneumocystis in vivo. Following immune reconstitution, alveolar macrophages from PcP-IRIS mice exhibited a dramatic increase in their ability to actively phagocytose Pneumocystis. Increased phagocytosis correlated temporally with fungal clearance, and required the presence of CD4(+) T cells. Sulfasalazine accelerated the onset of the CD4(+) T cell-dependent alveolar macrophage phagocytic response in PcP-IRIS mice, resulting in enhanced fungal clearance. Furthermore, sulfasalazine promoted a TH2-polarized cytokine environment in the lung, and sulfasalazine-enhanced phagocytosis of Pneumocystis was associated with an alternatively activated alveolar macrophage phenotype. These results provide evidence that macrophage phagocytosis is an important in vivo effector mechanism for T cell-mediated Pneumocystis clearance, and that macrophage phenotype can be altered to enhance phagocytosis without exacerbating inflammation. Immune modulation can diminish pulmonary inflammation while preserving host defense, and has therapeutic potential for the treatment of PcP-related immunopathogenesis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Immunomodulation/drug effects , Macrophages, Alveolar/drug effects , Pneumonia, Pneumocystis/immunology , Sulfasalazine/pharmacology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Separation/methods , Enzyme-Linked Immunosorbent Assay , Flow Cytometry/methods , Immune Reconstitution Inflammatory Syndrome/immunology , Immunomodulation/immunology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Mice , Mice, SCID , Microscopy, Confocal , Phagocytosis/drug effects , Reverse Transcriptase Polymerase Chain Reaction
7.
J Immunol ; 184(1): 497-502, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19949093

ABSTRACT

The T cell-mediated immune response elicited by Pneumocystis plays a key role in pulmonary damage and dysfunction during Pneumocystis carinii pneumonia (PcP). Mice depleted of CD4(+) and CD8(+) T cells prior to infection are markedly protected from PcP-related respiratory deficit and death, despite progressive lung infection. However, the therapeutic effectiveness of Ab-mediated disruption of T cell function in mice already displaying clinical symptoms of disease has not been determined. Therefore, a murine model of PcP-related immune reconstitution inflammatory syndrome was used to assess whether Ab to the pan-T cell molecule CD3 is effective for reducing the severity of PcP when administered after the onset of disease. Mice that received anti-CD3 Ab exhibited a rapid and dramatic halt in the PcP-associated pulmonary function decline within 1 week after treatment, and a striking enhancement of survival rate compared with mice receiving the control Ab. Physiologic improvement in anti-CD3 treated mice was associated with a significant reduction in the number of CD4(+) and CD8(+) T cells recovered in lung lavage fluid. This effectiveness of anti-CD3 was noted whether the mice also received antibiotic therapy with trimethoprim-sulfamethoxazole. These data suggest that monoclonal Ab-mediated disruption of T cell function may represent a specific and effective adjunctive therapy to rapidly reverse the ongoing pathologic immune response occurring during active PcP. Thus, the anti-human CD3 monoclonal Ab OKT3, which is already in clinical use, has the potential to be developed as an adjunctive therapy for PcP.


Subject(s)
Antibodies/therapeutic use , CD3 Complex/immunology , Inflammation/drug therapy , Lymphocyte Depletion/methods , Pneumonia, Pneumocystis/drug therapy , Animals , Anti-Infective Agents/therapeutic use , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Inflammation/immunology , Inflammation/microbiology , Mice , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/microbiology , Reverse Transcriptase Polymerase Chain Reaction , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use
8.
J Immunol ; 181(2): 1409-19, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606695

ABSTRACT

The opportunistic organism Pneumocystis carinii (Pc) produces a life-threatening pneumonia (PcP) in patients with low CD4(+) T cell counts. Animal models of HIV-AIDS-related PcP indicate that development of severe disease is dependent on the presence of CD8(+) T cells and the TNF receptors (TNFR) TNFRsf1a and TNFRsf1b. To distinguish roles of parenchymal and hematopoietic cell TNF signaling in PcP-related lung injury, murine bone marrow transplant chimeras of wild-type, C57BL6/J, and TNFRsf1a/1b double-null origin were generated, CD4(+) T cell depleted, and inoculated with Pc. As expected, C57 --> C57 chimeras (donor marrow --> recipient) developed significant disease as assessed by weight loss, impaired pulmonary function (lung resistance and dynamic lung compliance), and inflammatory cell infiltration. In contrast, TNFRsf1a/1b(-/-) --> TNFRsf1a/1b(-/-) mice were relatively mildly affected despite carrying the greatest organism burden. Mice solely lacking parenchymal TNFRs (C57 --> TNFRsf1a/1b(-/-)) had milder disease than did C57 --> C57 mice. Both groups of mice with TNFR-deficient parenchymal cells had low bronchoalveolar lavage fluid total cell counts and fewer lavageable CD8(+) T cells than did C57 --> C57 mice, suggesting that parenchymal TNFR signaling contributes to PcP-related immunopathology through the recruitment of damaging immune cells. Interestingly, mice with wild-type parenchymal cells but TNFRsf1a/1b(-/-) hematopoietic cells (TNFRsf1a/1b(-/-) --> C57) displayed exacerbated disease characterized by increased MCP-1 and KC production in the lung and increased macrophage and lymphocyte numbers in the lavage, indicating a dysregulated immune response. This study supports a key role of parenchymal cell TNFRs in lung injury induced by Pc and a potential protective effect of receptors on radiosensitive, bone marrow-derived cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Pneumonia, Pneumocystis/immunology , Receptors, Tumor Necrosis Factor/metabolism , Animals , Bone Marrow Transplantation , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , Hydro-Lyases/metabolism , Lung/immunology , Lung/microbiology , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumocystis carinii , Pneumonia, Pneumocystis/microbiology , Pneumonia, Pneumocystis/pathology , Pneumonia, Pneumocystis/physiopathology , Receptors, Tumor Necrosis Factor/immunology , Transplantation Chimera
9.
Am J Physiol Lung Cell Mol Physiol ; 292(6): L1495-505, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17307812

ABSTRACT

Pneumocystis carinii is an opportunistic fungal pathogen that causes pneumonia (PCP) in immunocompromised individuals. Recent studies have demonstrated that the host's immune response is clearly responsible for the majority of the pathophysiological changes associated with PCP. P. carinii interacts closely with alveolar epithelial cells (AECs); however, the nature and pathological consequences of the epithelial response remain poorly defined. Monocyte chemotactic protein-1 (MCP-1) is involved in lung inflammation, immunity, and epithelial repair and is upregulated during PCP. To determine whether AECs are an important source of MCP-1 in the P. carinii-infected lung, in vivo and in vitro studies were performed. In situ hybridization showed that MCP-1 mRNA was localized to cells with morphological characteristics of AECs in the lungs of infected mice. In vitro studies demonstrated that P. carinii stimulated a time- and dose-dependent MCP-1 response in primary murine type II cells that was preceded by JNK activation. Pharmacological inhibition of JNK nearly abolished P. carinii-stimulated MCP-1 production, while ERK, p38 MAPK, and TNF receptor signaling were not required. Furthermore, delivery of a JNK inhibitory peptide specifically to pulmonary epithelial cells using a recombinant adenovirus vector blocked the early lung MCP-1 response following intratracheal instillation of infectious P. carinii. JNK inhibition did not affect P. carinii-stimulated production of macrophage inflammatory protein-2 in vitro or in vivo, indicating that multiple signaling pathways are activated in P. carinii-stimulated AECs. These data demonstrate that AECs respond to P. carinii in a proinflammatory manner that may contribute to the generation of immune-mediated lung injury.


Subject(s)
Chemokine CCL2/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Pneumocystis Infections/metabolism , Pulmonary Alveoli/microbiology , Respiratory Mucosa/microbiology , Animals , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CXCL2 , Chemokines/metabolism , Disease Models, Animal , Epithelial Cells/enzymology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Gene Expression/immunology , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, SCID , NF-kappa B/metabolism , Pneumocystis Infections/immunology , Pneumocystis Infections/pathology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , beta-Glucans/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 291(6): L1256-66, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16891394

ABSTRACT

Immune-mediated lung injury is an important component of Pneumocystis pneumonia (PcP)-related immunorestitution disease (IRD). However, the individual contribution of CD4(+) and CD8(+) T cells to the pathophysiology of IRD remains undetermined. Therefore, IRD was modeled in severe combined immunodeficient mice, and specific T cell depletion was used to determine how T cell subsets interact to affect the nature and severity of disease. CD4(+) cells were more abundant than CD8(+) cells during the acute stage of IRD that coincided with impaired pulmonary physiology and organism clearance. Conversely, CD8(+) cells were more abundant during the resolution phase following P. carinii clearance. Depletion of CD4(+) T cells protected mice from the acute pathophysiology of IRD. However, these mice could not clear the infection and developed severe PcP at later time points when a pathological CD8(+) T cell response was observed. In contrast, mice depleted of CD8(+) T cells efficiently cleared the infection but developed more severe disease, an increased frequency of IFN-gamma-producing CD4(+) cells, and a prolonged CD4(+) T cell response than mice with both CD4(+) and CD8(+) cells. These data suggest that CD4(+) T cells mediate the acute respiratory disease associated with IRD. In contrast, CD8(+) T cells contributed to neither lung injury nor organism clearance when CD4(+) cells were present, but instead served to modulate CD4 function. In the absence of CD4(+) cells, CD8(+) T cells produced a nonprotective, pathological immune response. These data suggest that the interplay of CD4(+) and CD8(+) T cells affects the ultimate outcome of PcP-related IRD.


Subject(s)
Pneumocystis Infections/immunology , T-Lymphocyte Subsets/immunology , Animals , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , Female , Mice , Mice, SCID , Pneumocystis Infections/pathology , Pneumocystis carinii , Polymerase Chain Reaction , Serine Endopeptidases/genetics , T-Lymphocyte Subsets/pathology
11.
Infect Immun ; 74(11): 6310-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16940142

ABSTRACT

While CD8+ cells have been shown to contribute to lung injury during Pneumocystis carinii pneumonia (PCP), there are conflicting reports concerning the ability of CD8+ cells to kill P. carinii. To address these two issues, we studied the effect of the presence of CD8+ cells in two mouse models of PCP. In the reconstituted SCID mouse model, depletion of CD8+ cells in addition to CD4+ cells after reconstitution did not result in increased numbers of P. carinii cysts compared to the numbers of cysts in mice with only CD4+ cells depleted. This result was observed regardless of whether the mice were reconstituted with naïve or P. carinii-sensitized lymphocytes. In contrast, reconstitution with sensitized lymphocytes resulted in more rapid onset of lung injury that was dependent on the presence of CD8+ cells. The course of organism replication over a 6-week period was also examined in the CD4+-T-cell-depleted and CD4+- and CD8+-T-cell-depleted mouse model of PCP. Again, the organism burdens were identical at all times regardless of whether CD8+ cells were present. Thus, in the absence of CD4+ T cells, CD8+ T cells are a key contributor to the inflammatory lung injury associated with PCP. However, we were unable to demonstrate an in vivo effect of these cells on the course of P. carinii infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Pneumocystis carinii/immunology , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/microbiology , Animals , CD8-Positive T-Lymphocytes/pathology , Colony Count, Microbial , Disease Models, Animal , Disease Progression , Lymphocyte Depletion , Mice , Mice, SCID , Pneumonia, Pneumocystis/pathology , Spleen/cytology , Spleen/immunology , Spleen/transplantation
12.
Am J Physiol Lung Cell Mol Physiol ; 288(6): L1124-31, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15886399

ABSTRACT

This study examines the direct inhibitory effects of Pneumocystis carinii (Pc) organisms and chemical components on the surface activity and composition of whole calf lung surfactant (WLS) and calf lung surfactant extract (CLSE) in vitro. Incubation of WLS suspensions with intact Pc organisms (10(7) per milligram of surfactant phospholipid) did not significantly alter total phospholipid levels or surfactant protein A content. Incubation with intact Pc organisms also did not impair dynamic surface tension lowering in suspensions of WLS or centrifuged large surfactant aggregates on a bubble surfactometer (37 degrees C, 20 cycles/min, 0.5 and 2.5 mg phospholipid/ml). However, exposure of WLS or CLSE to disrupted (sonicated) Pc organisms led to severe detriments in activity, with minimum surface tensions of 17-19 mN/m vs. <1 mN/m for surfactants alone. Extracted hydrophobic chemical components from Pc (98.8% lipids, 0.1 mM) reduced the surface activity of WLS and CLSE similarly to sonicated Pc organisms, whereas extracted hydrophilic chemical components from Pc (primarily proteins) had only minor effects on surface tension lowering. These results indicate that in addition to surfactant dysfunction induced by inflammatory lung injury and edema-derived inhibitors in Pc pneumonia, disrupted Pc organisms in the alveolar lumen also have the potential to directly inhibit endogenous and exogenous lung surfactants in affected patients.


Subject(s)
Lung/metabolism , Phospholipids/metabolism , Pneumocystis carinii/pathogenicity , Pneumonia, Pneumocystis/metabolism , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactants/metabolism , Animals , Cattle , In Vitro Techniques , Lung/pathology , Mice , Mice, SCID , Pneumonia, Pneumocystis/immunology , Surface Tension
SELECTION OF CITATIONS
SEARCH DETAIL
...