Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
bioRxiv ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38746361

ABSTRACT

RATIONALE: Asthma is a chronic inflammatory disease of the airways that involves crosstalk between myeloid-derived regulatory cells (MDRCs) and CD4+ T cells. Although small extracellular vesicles (sEVs) are known to mediate cell-cell communication, the role of sEV signaling via mitochondria in perpetuating asthmatic airway inflammation is unknown. OBJECTIVES: We investigated the effects of MDRC-derived exosomes on dysregulated T cell responses in asthmatics. METHODS: Small extracellular vesicles isolated from bronchoalveolar lavage fluid or airway MDRCs of mild to moderate asthmatics or healthy controls were co-cultured with autologous peripheral and airway CD4+ T lymphocytes. sEV internalization, sEV-mediated transfer of mitochondria targeted GFP to T cells, sEV mitochondrial signaling, and subsequent activation, proliferation and polarization of CD4+ T lymphocytes to Th1, Th2 and Th17 subsets were assessed. MEASUREMENTS AND MAIN RESULTS: Airway MDRC-derived sEVs from asthmatics mediated T cell receptor engagement and transfer of mitochondria that induced antigen-specific activation and polarization into Th17 and Th2 cells, drivers of chronic airway inflammation in asthma. CD4+ T cells internalized sEVs containing mitochondria predominantly by membrane fusion, and blocking mitochondrial oxidant signaling in MDRC-derived exosomes mitigated T cell activation. Reactive oxygen species-mediated signaling that elicited T cell activation in asthmatics was sEV-dependent. A Drp1-dependent mitochondrial fission in pro-inflammatory MDRCs promoted mitochondrial packaging within sEVs, which then co-localized with the polarized actin cytoskeleton and mitochondrial networks in the organized immune synapse of recipient T cells. CONCLUSIONS: Our studies indicate a previously unrecognized role for mitochondrial fission and exosomal mitochondrial transfer in dysregulated T cell activation and Th cell differentiation in asthma which could constitute a novel therapeutic target.

2.
Sci Transl Med ; 10(455)2018 08 22.
Article in English | MEDLINE | ID: mdl-30135247

ABSTRACT

It is anticipated that bioactive fragments of the extracellular matrix (matrikines) can influence the development and progression of chronic diseases. The enzyme leukotriene A4 hydrolase (LTA4H) mediates opposing proinflammatory and anti-inflammatory activities, through the generation of leukotriene B4 (LTB4) and degradation of proneutrophilic matrikine Pro-Gly-Pro (PGP), respectively. We show that abrogation of LTB4 signaling ameliorated inflammation and airway hyperresponsiveness (AHR) in a murine asthma model, yet global loss of LTA4H exacerbated AHR, despite the absence of LTB4 This exacerbated AHR was attributable to a neutrophil-independent capacity of PGP to promote pathological airway epithelial remodeling. Thus, we demonstrate a disconnect between airway inflammation and AHR and the ability of a matrikine to promote an epithelial remodeling phenotype that negatively affects lung function. Subsequently, we show that substantial quantities of PGP are detectable in the sputum of moderate-severe asthmatics in two distinct cohorts of patients. These studies have implications for our understanding of remodeling phenotypes in asthma and may rationalize the failure of LTA4H inhibitors in the clinic.


Subject(s)
Airway Remodeling , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Respiratory Hypersensitivity/physiopathology , Airway Resistance , Animals , Asthma/complications , Asthma/immunology , Asthma/pathology , Asthma/physiopathology , Bronchi/pathology , Cell Count , Disease Models, Animal , Epoxide Hydrolases/deficiency , Epoxide Hydrolases/metabolism , Humans , Hypersensitivity/complications , Hypersensitivity/immunology , Hypersensitivity/pathology , Hypersensitivity/physiopathology , Inflammation/pathology , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mucus/metabolism , Neutrophils/metabolism , Oligopeptides/metabolism , Proline/analogs & derivatives , Proline/metabolism , Pyroglyphidae/physiology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/parasitology , Respiratory Hypersensitivity/pathology , Sputum/metabolism , T-Lymphocytes, Helper-Inducer/immunology
3.
Sci Rep ; 8(1): 10340, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29985427

ABSTRACT

Asthma is a chronic inflammatory disease process involving the conductive airways of the human lung. The dysregulated inflammatory response in this disease process may involve multiple cell-cell interactions mediated by signaling molecules, including lipid mediators. Extracellular vesicles (EVs) are lipid membrane particles that are now recognized as critical mediators of cell-cell communication. Here, we compared the lipid composition and presence of specific lipid mediators in airway EVs purified from the bronchoalveolar lavage (BAL) fluid of healthy controls and asthmatic subjects with and without second-hand smoke (SHS) exposure. Airway exosome concentrations were increased in asthmatics, and correlated with blood eosinophilia and serum IgE levels. Frequencies of HLA-DR+ and CD54+ exosomes were also significantly higher in asthmatics. Lipidomics analysis revealed that phosphatidylglycerol, ceramide-phosphates, and ceramides were significantly reduced in exosomes from asthmatics compared to the non-exposed control groups. Sphingomyelin 34:1 was more abundant in exosomes of SHS-exposed asthmatics compared to healthy controls. Our results suggest that chronic airway inflammation may be driven by alterations in the composition of lipid mediators within airway EVs of human subjects with asthma.


Subject(s)
Asthma/pathology , Extracellular Vesicles/metabolism , Adult , Aged , Bronchoalveolar Lavage Fluid/chemistry , Case-Control Studies , Ceramides/metabolism , Discriminant Analysis , Down-Regulation , Exosomes/metabolism , Female , HLA-DR Antigens/metabolism , Humans , Immunoglobulin E/blood , Intercellular Adhesion Molecule-1/metabolism , Male , Middle Aged , Phosphatidylglycerols/metabolism , Sphingomyelins/metabolism , Tobacco Smoke Pollution
4.
Redox Biol ; 18: 54-64, 2018 09.
Article in English | MEDLINE | ID: mdl-29986209

ABSTRACT

Chronic inflammation involving both innate and adaptive immune cells is implicated in the pathogenesis of asthma. Intercellular communication is essential for driving and resolving inflammatory responses in asthma. Emerging studies suggest that extracellular vesicles (EVs) including exosomes facilitate this process. In this report, we have used a range of approaches to show that EVs contain markers of mitochondria derived from donor cells which are capable of sustaining a membrane potential. Further, we propose that these participate in intercellular communication within the airways of human subjects with asthma. Bronchoalveolar lavage fluid of both healthy volunteers and asthmatics contain EVs with encapsulated mitochondria; however, the % HLA-DR+ EVs containing mitochondria and the levels of mitochondrial DNA within EVs were significantly higher in asthmatics. Furthermore, mitochondria are present in exosomes derived from the pro-inflammatory HLA-DR+ subsets of airway myeloid-derived regulatory cells (MDRCs), which are known regulators of T cell responses in asthma. Exosomes tagged with MitoTracker Green, or derived from MDRCs transduced with CellLight Mitochondrial GFP were found in recipient peripheral T cells using a co-culture system, supporting direct exosome-mediated cell-cell transfer. Importantly, exosomally transferred mitochondria co-localize with the mitochondrial network and generate reactive oxygen species within recipient T cells. These findings support a potential novel mechanism of cell-cell communication involving exosomal transfer of mitochondria and the bioenergetic and/or redox regulation of target cells.


Subject(s)
Asthma/pathology , Exosomes/pathology , Mitochondria/pathology , Myeloid Cells/pathology , Cell Communication , DNA, Mitochondrial/analysis , HLA-DR Antigens/analysis , Humans , Oxidation-Reduction , Reactive Oxygen Species/analysis
5.
Am J Med ; 131(5): 484-491, 2018 05.
Article in English | MEDLINE | ID: mdl-29355509

ABSTRACT

Most patients with asthma are managed by primary care providers. Severe asthma is associated with substantial morbidity and health care resource use, and long-term sequelae of severe asthma include airway remodeling and a greater risk of developing chronic obstructive pulmonary disease. These consequences highlight the importance of early identification and improved management of patients with severe asthma. Although treatment guidelines can be confusing and it can be difficult to keep abreast of updates, routine assessments of lung function, frequency and severity of exacerbations, symptom control, and medication adherence in the primary care setting provide the necessary information for identifying severe asthma and determining appropriate management strategies. An increased understanding of asthma pathophysiology and its relationship to disease activity has identified therapeutic targets and associated biomarkers. Biologic therapies directed at these targets offer individualized targeted treatment of severe asthma. We review evidence-based guidelines for identification and management of severe asthma, clarify the relationship of asthma control and asthma severity, and provide an overview of new biologic therapies offering additional treatment options for patients with severe asthma.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/diagnosis , Asthma/drug therapy , Primary Health Care , Anti-Asthmatic Agents/pharmacology , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cytokines/physiology , Eosinophilia/diagnosis , Humans , Immunoglobulin E/blood , Nitric Oxide/metabolism , Practice Guidelines as Topic , Severity of Illness Index
7.
N Engl J Med ; 376(20): 1911-1920, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28514613

ABSTRACT

BACKGROUND: Mast cells are present in the airways of patients who have severe asthma despite glucocorticoid treatment; these cells are associated with disease characteristics including poor quality of life and inadequate asthma control. Stem cell factor and its receptor, KIT, are central to mast-cell homeostasis. We conducted a proof-of-principle trial to evaluate the effect of imatinib, a KIT inhibitor, on airway hyperresponsiveness, a physiological marker of severe asthma, as well as on airway mast-cell numbers and activation in patients with severe asthma. METHODS: We conducted a randomized, double-blind, placebo-controlled, 24-week trial of imatinib in patients with poorly controlled severe asthma who had airway hyperresponsiveness despite receiving maximal medical therapy. The primary end point was the change in airway hyperresponsiveness, measured as the concentration of methacholine required to decrease the forced expiratory volume in 1 second by 20% (PC20). Patients also underwent bronchoscopy. RESULTS: Among the 62 patients who underwent randomization, imatinib treatment reduced airway hyperresponsiveness to a greater extent than did placebo. At 6 months, the methacholine PC20 increased by a mean (±SD) of 1.73±0.60 doubling doses in the imatinib group, as compared with 1.07±0.60 doubling doses in the placebo group (P=0.048). Imatinib also reduced levels of serum tryptase, a marker of mast-cell activation, to a greater extent than did placebo (decrease of 2.02±2.32 vs. 0.56±1.39 ng per milliliter, P=0.02). Airway mast-cell counts declined in both groups. Muscle cramps and hypophosphatemia were more common in the imatinib group than in the placebo group. CONCLUSIONS: In patients with severe asthma, imatinib decreased airway hyperresponsiveness, mast-cell counts, and tryptase release. These results suggest that KIT-dependent processes and mast cells contribute to the pathobiologic basis of severe asthma. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT01097694 .).


Subject(s)
Asthma/drug therapy , Imatinib Mesylate/therapeutic use , Mast Cells/metabolism , Protein Kinase Inhibitors/therapeutic use , Adult , Asthma/immunology , Asthma/physiopathology , Bronchial Hyperreactivity/drug therapy , Bronchial Provocation Tests , Cell Count , Double-Blind Method , Female , Forced Expiratory Volume/drug effects , Humans , Imatinib Mesylate/adverse effects , Male , Methacholine Chloride , Middle Aged , Protein Kinase Inhibitors/adverse effects , Quality of Life , Tryptases/blood , Tryptases/metabolism
8.
Lancet Respir Med ; 5(5): 390-400, 2017 05.
Article in English | MEDLINE | ID: mdl-28395936

ABSTRACT

BACKGROUND: Mepolizumab, an anti-interleukin-5 monoclonal antibody approved as add-on therapy to standard of care for patients with severe eosinophilic asthma, has been shown in previous studies to reduce exacerbations and dependency on oral corticosteroids compared with placebo. We aimed to further assess mepolizumab in patients with severe eosinophilic asthma by examining its effect on health-related quality of life (HRQOL). METHODS: We did a randomised, double-blind, placebo-controlled, parallel-group, multicentre, phase 3b trial (MUSCA) in 146 hospitals or research centres in 19 countries worldwide. Eligible participants were patients aged 12 years or older with severe eosinophilic asthma and a history of at least two exacerbations requiring treatment in the previous 12 months before screening despite regular use of high-dose inhaled corticosteroids plus other controller medicines. Exclusion criteria included current smokers or former smokers with a history of at least ten pack-years. We randomly assigned participants (1:1) by country to receive a subcutaneous injection of either mepolizumab 100 mg or placebo, plus standard of care, every 4 weeks for 24 weeks (the final dose was given at week 20). We did the randomisation using an interactive voice response system and a centralised, computer-generated, permuted-block design of block size six. The two treatments were identical in appearance and administered in a masked manner; patients, investigators, other site staff and the entire study team including those assessing outcomes data were also masked to group assignment. The primary endpoint was the mean change from baseline in the St George's Respiratory Questionnaire (SGRQ) total score at week 24 in the modified intention-to-treat (modified ITT) population (analysed according to their randomly assigned treatment). Safety was assessed in all patients who received at least one dose of trial medication (analysed according to the actual treatment received). This trial is registered with ClinicalTrials.gov, number NCT02281318. FINDINGS: We recruited patients between Dec 11, 2014, and Nov 20, 2015, and the study was undertaken between Dec 11, 2014, and June 10, 2016. The modified ITT population comprised 274 patients assigned to mepolizumab 100 mg and 277 assigned to placebo. Mepolizumab versus placebo showed significant improvements at week 24 from baseline in SGRQ total score (least squares mean [SE] change from baseline -15·6 (1·0) vs -7·9 (1·0), a treatment difference of -7·7 (95% CI -10·5 to -4·9; p<0·0001). No deaths occurred during the study. 192 (70%) of 273 patients who received mepolizumab and 207 (74%) of 278 who received placebo reported at least one on-treatment adverse event, the most common of which were headache (in 45 [16%] given mepolizumab vs 59 [21%] given placebo) and nasopharyngitis (in 31 [11%] given mepolizumab vs 46 [17%] given placebo). 15 (5%) and 22 (8%) patients had an on-treatment serious adverse event in the mepolizumab and placebo groups, respectively; the most common was asthma in both groups (in three [1%] given mepolizumab vs nine [3%] given placebo). INTERPRETATION: Mepolizumab was associated with significant improvements in HRQOL in patients with severe eosinophilic asthma, and had a safety profile similar to that of placebo. These results add to and support the use of mepolizumab as a favourable add-on treatment option to standard of care in patients with severe eosinophilic asthma. FUNDING: GlaxoSmithKline.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/drug therapy , Eosinophilia/drug therapy , Quality of Life , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Asthma/complications , Bronchodilator Agents/therapeutic use , Disease Progression , Double-Blind Method , Drug Therapy, Combination , Eosinophilia/complications , Female , Headache/chemically induced , Humans , Male , Middle Aged , Nasopharyngitis/chemically induced , Severity of Illness Index , Symptom Assessment , Symptom Flare Up
9.
Ann Am Thorac Soc ; 13(2): 197-203, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26599286

ABSTRACT

RATIONALE: Hospitalization for acute exacerbation of chronic obstructive pulmonary disease (COPD) is associated with significant morbidity and health care costs, and hospitals in the United States are now penalized by the Centers for Medicare and Medicaid Services for excessive readmissions. Identifying patients at risk of readmission is important, but modifiable risk factors have not been clearly established, and the potential contributing role of psychological disease has not been examined adequately. We hypothesized that depression and anxiety would increase the risk of both short- and long-term readmissions for acute exacerbation of COPD. OBJECTIVES: To characterize the associations between depression and anxiety and COPD readmission risk. METHODS: We examined the medical records for all patients with a primary diagnosis of acute exacerbation of COPD by International Classification of Diseases, Ninth Revision codes admitted to the University of Alabama at Birmingham Hospital between November 2010 and October 2012. Those who did not meet the standardized study criteria for acute exacerbation of COPD and those with other respiratory illnesses as the primary diagnosis were excluded. Comorbidities were recorded on the basis of physician documentation of the diagnosis and/or the use of medications in the electronic medical record. Multivariable regression analyses identified factors associated with readmission for acute exacerbation of COPD at 1 year and within 30 and 90 days. MEASUREMENTS AND MAIN RESULTS: Four hundred twenty-two patients were included, with 132 readmitted in 1 year. Mean age was 64.8 ± 11.7 years, and mean percent predicted FEV1 was 48.1 ± 18.7%. On univariate analysis, readmitted patients had lower percent predicted FEV1 (44.9 ± 17.3% vs. 50.2 ± 19.4%; P = 0.05) and a higher frequency of depression (47.7% vs. 23.4%; P < 0.001). On multivariable analysis, 1-year readmission was independently associated with depression (adjusted odds ratio [OR], 2.67; 95% confidence interval [CI], 1.59-4.47) and in-hospital tobacco cessation counseling (adjusted OR, 0.34; 95% CI, 0.18-0.66). Depression also predicted readmission at 30 days (adjusted OR, 3.83; 95% CI, 1.84-7.96) and 90 days (adjusted OR, 2.47; 95% CI, 1.34-4.55). CONCLUSIONS: Depression is an independent risk factor for both short- and long-term readmissions for acute exacerbation of COPD and may represent a modifiable risk factor. In-hospital tobacco cessation counseling was also associated with reduced 1-year readmission.


Subject(s)
Depression/epidemiology , Depressive Disorder/epidemiology , Patient Readmission/statistics & numerical data , Pulmonary Disease, Chronic Obstructive/epidemiology , Aged , Cohort Studies , Comorbidity , Counseling/statistics & numerical data , Disease Progression , Female , Hospitalization , Humans , Logistic Models , Male , Middle Aged , Multivariate Analysis , Pulmonary Disease, Chronic Obstructive/psychology , Pulmonary Disease, Chronic Obstructive/therapy , Retrospective Studies , Risk Factors , Smoking/epidemiology , Smoking/therapy , Smoking Cessation/statistics & numerical data , United States/epidemiology
10.
J Asthma ; 52(9): 969-73, 2015.
Article in English | MEDLINE | ID: mdl-26287942

ABSTRACT

BACKGROUND: Asthma affects 30 million Americans and results in reduced productivity and quality of life. Pulmonary rehabilitation (PR) is known to improve physical conditioning and exercise performance in chronic lung diseases such as COPD, however, few studies have examined its benefits in patients with asthma. We aimed to determine the benefits of PR in this population as well as the predictors of completion of therapy. METHODS: We performed a retrospective review of data from patients with a diagnosis of asthma who participated in PR at our institution from 1996 to 2013. Nine hundred and nineteen patients participated in the program of whom 75 were referred with a primary diagnosis of asthma. Patients underwent physiologic testing and their symptoms and quality of life were assessed using validated questionnaires. For patients who completed PR (n = 37), data obtained at the initial and exit visit was compared. Characteristics of completers were compared to non-completers to determine predictors of successful completion. RESULTS: Individuals with asthma completing PR had improvement from baseline to exit visit in Six Minute Walk Distance (326 vs. 390 feet; p < 0.0001), decreased body mass index (33 vs. 32 kg/m2; p < 0.046), decreased Beck Depression Inventory scores (15 vs. 9; p < 0.0009), and increased Short Form-36 scores (345 vs. 445; p = 0.0005). In a multivariate logistic regression analysis, lower depression scores predicted completion (OR 1.08, 95% CI 1.02-1.15, p = 0.02). CONCLUSION: Patients with asthma who completed PR had improvement in physical function and emotional well-being. Depression is a risk factor for non-completion of PR. Further research is needed to determine which patients will benefit most from therapy.


Subject(s)
Asthma/rehabilitation , Quality of Life , Adult , Aged , Asthma/psychology , Depression/psychology , Exercise Tolerance , Female , Humans , Male , Middle Aged , Retrospective Studies
11.
J Immunol ; 190(12): 6287-94, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23650614

ABSTRACT

Although it is clear that the loss of CD4(+) T cells is a predisposing factor for the development of Pneumocystis pneumonia, specific Th mechanisms mediating protection are not well understood. Th1, Th2, and Th17 responses have each been implicated in protective responses during infection. As STAT4 may promote Th1 and Th17 development, yet antagonize Th2 development, we investigated its role in Pneumocystis murina host defense. STAT4 was required for Th1 and, unexpectedly, Th2 responses in the lungs of C57BL/6 (BL/6) and BALB/c mice 14 d postchallenge, but only BALB/c Stat4(-/-) mice demonstrated susceptibility to P. murina lung infection. BL/6 Stat4(-/-), but not BALB/c Stat4(-/-), mice maintained an enhanced alternatively activated (M2) macrophage signature in the lungs, which we have previously reported to be associated with enhanced P. murina clearance. In addition, anti-P. murina class-switched Abs were increased in BL/6 Stat4(-/-) mice, but not BALB/c Stat4(-/-) mice. Supporting our experimental observations, plasma from HIV-infected individuals colonized with Pneumocystis jirovecii contained significantly lower levels of the Th2 cytokines IL-4, IL-5, and IL-13 compared with HIV-infected individuals who were not colonized. Collectively, our data suggest that robust local and systemic Th2-mediated responses are critical for immunity to Pneumocystis.


Subject(s)
HIV Infections/complications , Pneumonia, Pneumocystis/immunology , STAT4 Transcription Factor/immunology , Th2 Cells/immunology , Animals , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , HIV Infections/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Pneumocystis/complications , Real-Time Polymerase Chain Reaction
12.
J Immunol ; 186(4): 2372-81, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21220696

ABSTRACT

We have recently reported that mice deficient in the myeloid Src-family tyrosine kinases Hck, Fgr, and Lyn (Src triple knockout [TKO]) had augmented innate lung clearance of Pneumocystis murina that correlated with a higher ability of alveolar macrophages (AMs) from these mice to kill P. murina. In this article, we show that despite possessing enhanced killing, AMs from naive Src TKO mice did not demonstrate enhanced inflammatory responses to P. murina. We subsequently discovered that both AMs and lungs from P. murina-infected Src TKO mice expressed significantly greater levels of the M2a markers RELM-α and Arg1, and the M2a-associated chemokines CCL17 and CCL22 than did wild-type mice. IL-4 and IL-13, the primary cytokines that promote M2a polarization, were not differentially produced in the lungs between wild-type and Src TKO mice. P. murina infection in Src TKO mice resulted in enhanced lung production of the novel IL-1 family cytokine IL-33. Immunohistochemical analysis of IL-33 in lung tissue revealed localization predominantly in the nucleus of alveolar epithelial cells. We further demonstrate that experimental polarization of naive AMs to M2a resulted in more efficient killing of P. murina compared with untreated AMs, which was further enhanced by the addition of IL-33. Administration of IL-33 to C57BL/6 mice increased lung RELM-α and CCL17 levels, and enhanced clearance of P. murina, despite having no effect on the cellular composition of the lungs. Collectively, these results indicate that M2a AMs are potent effector cells against P. murina. Furthermore, enhancing M2a polarization may be an adjunctive therapy for the treatment of Pneumocystis.


Subject(s)
Interleukins/physiology , Macrophages, Alveolar/immunology , Pneumocystis/immunology , Pneumocystis/pathogenicity , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/prevention & control , Animals , Cell Polarity/genetics , Cell Polarity/immunology , Cells, Cultured , Immunity, Innate/genetics , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Interleukin-33 , Interleukins/biosynthesis , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/genetics , Phagocytosis/immunology , Pneumocystis/growth & development , Pneumonia, Pneumocystis/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-hck/deficiency , Proto-Oncogene Proteins c-hck/physiology , src-Family Kinases/deficiency , src-Family Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...