Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 326(1): L39-L51, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37933452

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is marked by unremitting matrix deposition and architectural distortion. Multiple profibrotic pathways contribute to the persistent activation of mesenchymal cells (MCs) in fibrosis, highlighting the need to identify and target common signaling pathways. The transcription factor nuclear factor of activated T cells 1 (NFAT1) lies downstream of second messenger calcium signaling and has been recently shown to regulate key profibrotic mediator autotaxin (ATX) in lung MCs. Herein, we investigate the role of NFAT1 in regulating fibroproliferative responses during the development of lung fibrosis. Nfat1-/--deficient mice subjected to bleomycin injury demonstrated improved survival and protection from lung fibrosis and collagen deposition as compared with bleomycin-injured wild-type (WT) mice. Chimera mice, generated by reconstituting bone marrow cells from WT or Nfat1-/- mice into irradiated WT mice (WT→WT and Nfat1-/-→WT), demonstrated no difference in bleomycin-induced fibrosis, suggesting immune influx-independent fibroprotection in Nfat1-/- mice. Examination of lung tissue and flow sorted lineageneg/platelet-derived growth factor receptor alpha (PDGFRα)pos MCs demonstrated decreased MC numbers, proliferation [↓ cyclin D1 and 5-ethynyl-2'-deoxyuridine (EdU) incorporation], myofibroblast differentiation [↓ α-smooth muscle actin (α-SMA)], and survival (↓ Birc5) in Nfat1-/- mice. Nfat1 deficiency abrogated ATX expression in response to bleomycin in vivo and MCs derived from Nfat1-/- mice demonstrated decreased ATX expression and migration in vitro. Human IPF MCs demonstrated constitutive NFAT1 activation, and regulation of ATX in these cells by NFAT1 was confirmed using pharmacological and genetic inhibition. Our findings identify NFAT1 as a critical mediator of profibrotic processes, contributing to dysregulated lung remodeling and suggest its targeting in MCs as a potential therapeutic strategy in IPF.NEW & NOTEWORTHY Idiopathic pulmonary fibrosis (IPF) is a fatal disease with hallmarks of fibroblastic foci and exuberant matrix deposition, unknown etiology, and ineffective therapies. Several profibrotic/proinflammatory pathways are implicated in accelerating tissue remodeling toward a honeycombed end-stage disease. NFAT1 is a transcriptional factor activated in IPF tissues. Nfat1-deficient mice subjected to chronic injury are protected against fibrosis independent of immune influxes, with suppression of profibrotic mesenchymal phenotypes including proliferation, differentiation, resistance to apoptosis, and autotaxin-related migration.


Asunto(s)
Fibrosis Pulmonar Idiopática , Pulmón , Animales , Humanos , Ratones , Bleomicina/farmacología , Diferenciación Celular/genética , Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal
2.
EMBO J ; 39(16): e105057, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32643835

RESUMEN

Alveolar macrophages (AMs) and epithelial cells (ECs) are the lone resident lung cells positioned to respond to pathogens at early stages of infection. Extracellular vesicles (EVs) are important vectors of paracrine signaling implicated in a range of (patho)physiologic contexts. Here we demonstrate that AMs, but not ECs, constitutively secrete paracrine activity localized to EVs which inhibits influenza infection of ECs in vitro and in vivo. AMs exposed to cigarette smoke extract lost the inhibitory activity of their secreted EVs. Influenza strains varied in their susceptibility to inhibition by AM-EVs. Only those exhibiting early endosomal escape and high pH of fusion were inhibited via a reduction in endosomal pH. By contrast, strains exhibiting later endosomal escape and lower fusion pH proved resistant to inhibition. These results extend our understanding of how resident AMs participate in host defense and have broader implications in the defense and treatment of pathogens internalized within endosomes.


Asunto(s)
Endosomas , Vesículas Extracelulares/inmunología , Virus de la Influenza A/inmunología , Macrófagos Alveolares/inmunología , Comunicación Paracrina/inmunología , Internalización del Virus , Células A549 , Animales , Perros , Endosomas/inmunología , Endosomas/patología , Endosomas/virología , Células HEK293 , Humanos , Macrófagos Alveolares/patología , Células de Riñón Canino Madin Darby , Ratones , Ratas , Ratas Wistar , Células THP-1
3.
PLoS Pathog ; 15(1): e1007560, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30682165

RESUMEN

Bacterial lung infections, particularly with methicillin-resistant Staphylococcus aureus (MRSA), increase mortality following influenza infection, but the mechanisms remain unclear. Here we show that expression of TLR9, a microbial DNA sensor, is increased in murine lung macrophages, dendritic cells, CD8+ T cells and epithelial cells post-influenza infection. TLR9-/- mice did not show differences in handling influenza nor MRSA infection alone. However, TLR9-/- mice have improved survival and bacterial clearance in the lung post-influenza and MRSA dual infection, with no difference in viral load during dual infection. We demonstrate that TLR9 is upregulated on macrophages even when they are not themselves infected, suggesting that TLR9 upregulation is related to soluble mediators. We rule out a role for elevations in interferon-γ (IFNγ) in mediating the beneficial MRSA clearance in TLR9-/- mice. While macrophages from WT and TLR9-/- mice show similar phagocytosis and bacterial killing to MRSA alone, following influenza infection, there is a marked upregulation of scavenger receptor A and MRSA phagocytosis as well as inducible nitric oxide synthase (Inos) and improved bacterial killing that is specific to TLR9-deficient cells. Bone marrow transplant chimera experiments and in vitro experiments using TLR9 antagonists suggest TLR9 expression on non-hematopoietic cells, rather than the macrophages themselves, is important for regulating myeloid cell function. Interestingly, improved bacterial clearance post-dual infection was restricted to MRSA, as there was no difference in the clearance of Streptococcus pneumoniae. Taken together these data show a surprising inhibitory role for TLR9 signaling in mediating clearance of MRSA that manifests following influenza infection.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina/inmunología , Staphylococcus aureus Resistente a Meticilina/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Humanos , Gripe Humana/inmunología , Pulmón/inmunología , Macrófagos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Fagocitosis , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Receptor Toll-Like 9/genética
4.
Arterioscler Thromb Vasc Biol ; 39(11): 2353-2366, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31644352

RESUMEN

OBJECTIVE: Sepsis represents an acute life-threatening disorder resulting from a dysregulated host response. For patients who survive sepsis, there remains long-term consequences, including impaired inflammation, as a result of profound immunosuppression. The mechanisms involved in this long-lasting deficient immune response are poorly defined. Approach and Results: Sepsis was induced using the murine model of cecal ligation and puncture. Following a full recovery period from sepsis physiology, mice were subjected to our wound healing model and wound macrophages (CD11b+, CD3-, CD19-, Ly6G-) were sorted. Post-sepsis mice demonstrated impaired wound healing and decreased reepithelization in comparison to controls. Further, post-sepsis bone marrow-derived macrophages and wound macrophages exhibited decreased expression of inflammatory cytokines vital for wound repair (IL [interleukin]-1ß, IL-12, and IL-23). To evaluate if decreased inflammatory gene expression was secondary to epigenetic modification, we conducted chromatin immunoprecipitation on post-sepsis bone marrow-derived macrophages and wound macrophages. This demonstrated decreased expression of Mll1, an epigenetic enzyme, and impaired histone 3 lysine 4 trimethylation (activation mark) at NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells)-binding sites on inflammatory gene promoters in bone marrow-derived macrophages and wound macrophages from postcecal ligation and puncture mice. Bone marrow transplantation studies demonstrated epigenetic modifications initiate in bone marrow progenitor/stem cells following sepsis resulting in lasting impairment in peripheral macrophage function. Importantly, human peripheral blood leukocytes from post-septic patients demonstrate a significant reduction in MLL1 compared with nonseptic controls. CONCLUSIONS: These data demonstrate that severe sepsis induces stable mixed-lineage leukemia 1-mediated epigenetic modifications in the bone marrow, which are passed to peripheral macrophages resulting in impaired macrophage function and deficient wound healing persisting long after sepsis recovery.


Asunto(s)
Epigénesis Genética , Inflamación/fisiopatología , Macrófagos/fisiología , Sepsis/genética , Sepsis/fisiopatología , Cicatrización de Heridas/fisiología , Animales , Células de la Médula Ósea/fisiología , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Humanos , Tolerancia Inmunológica , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos , Proteína de la Leucemia Mieloide-Linfoide/genética , FN-kappa B/genética , Regiones Promotoras Genéticas , Sepsis/metabolismo
5.
Am J Respir Crit Care Med ; 200(1): 63-74, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30742492

RESUMEN

Rationale: "Noninfectious" pulmonary complications are significant causes of morbidity and mortality after allogeneic hematopoietic cell transplant. Early-onset viral reactivations or infections are common after transplant. Whether the first-onset viral infection causes noninfectious pulmonary complications is unknown. Objectives: To determine whether the first-onset viral infection within 100 days after transplant predisposes to development of noninfectious pulmonary complications. Methods: We performed a retrospective review of 738 allogeneic hematopoietic cell transplant patients enrolled from 2005 to 2011. We also established a novel bone marrow transplantation mouse model to test whether herpesviral reactivation after transplant causes organ injury. Measurements and Main Results: First-onset viral infections with human herpesvirus 6 or Epstein-Barr virus within 100 days after transplant increase the risk of developing idiopathic pneumonia syndrome (adjusted hazard ratio [aHR], 5.52; 95% confidence interval [CI], 1.61-18.96; P = 0.007; and aHR, 9.21; 95% CI, 2.63-32.18; P = 0.001, respectively). First infection with human cytomegalovirus increases risk of bronchiolitis obliterans syndrome (aHR, 2.88; 95% CI, 1.50-5.55; P = 0.002) and grade II-IV acute graft-versus-host disease (aHR, 1.59; 95% CI, 1.06-2.39; P = 0.02). Murine roseolovirus, a homolog of human herpesvirus 6, can also be reactivated in the lung and other organs after bone marrow transplantation. Reactivation of murine roseolovirus induced an idiopathic pneumonia syndrome-like phenotype and aggravated acute graft-versus-host disease. Conclusions: First-onset herpesviral infection within 100 days after allogeneic hematopoietic cell transplant increases risk of pulmonary complications. Experimentally reactivating murine roseolovirus causes organ injury similar to phenotypes seen in human transplant recipients.


Asunto(s)
Bronquiolitis Obliterante/epidemiología , Enfermedad Injerto contra Huésped/epidemiología , Trasplante de Células Madre Hematopoyéticas , Infecciones por Herpesviridae/epidemiología , Lesión Pulmonar/epidemiología , Neumonía/epidemiología , Complicaciones Posoperatorias/epidemiología , Trasplante Homólogo , Adolescente , Adulto , Anciano , Animales , Niño , Preescolar , Infecciones por Citomegalovirus/epidemiología , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/epidemiología , Femenino , Herpes Simple/epidemiología , Humanos , Lactante , Masculino , Ratones , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Infecciones por Roseolovirus/epidemiología , Activación Viral , Adulto Joven
6.
Am J Respir Crit Care Med ; 199(9): 1127-1138, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30789747

RESUMEN

Rationale: Idiopathic pulmonary fibrosis (IPF) causes considerable global morbidity and mortality, and its mechanisms of disease progression are poorly understood. Recent observational studies have reported associations between lung dysbiosis, mortality, and altered host defense gene expression, supporting a role for lung microbiota in IPF. However, the causal significance of altered lung microbiota in disease progression is undetermined. Objectives: To examine the effect of microbiota on local alveolar inflammation and disease progression using both animal models and human subjects with IPF. Methods: For human studies, we characterized lung microbiota in BAL fluid from 68 patients with IPF. For animal modeling, we used a murine model of pulmonary fibrosis in conventional and germ-free mice. Lung bacteria were characterized using 16S rRNA gene sequencing with novel techniques optimized for low-biomass sample load. Microbiota were correlated with alveolar inflammation, measures of pulmonary fibrosis, and disease progression. Measurements and Main Results: Disruption of the lung microbiome predicts disease progression, correlates with local host inflammation, and participates in disease progression. In patients with IPF, lung bacterial burden predicts fibrosis progression, and microbiota diversity and composition correlate with increased alveolar profibrotic cytokines. In murine models of fibrosis, lung dysbiosis precedes peak lung injury and is persistent. In germ-free animals, the absence of a microbiome protects against mortality. Conclusions: Our results demonstrate that lung microbiota contribute to the progression of IPF. We provide biological plausibility for the hypothesis that lung dysbiosis promotes alveolar inflammation and aberrant repair. Manipulation of lung microbiota may represent a novel target for the treatment of IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática/microbiología , Inflamación/microbiología , Pulmón/microbiología , Microbiota/fisiología , Anciano , Animales , Líquido del Lavado Bronquioalveolar/microbiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Vida Libre de Gérmenes , Humanos , Fibrosis Pulmonar Idiopática/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota/genética , Persona de Mediana Edad , Alveolos Pulmonares/microbiología , Alveolos Pulmonares/patología , ARN Ribosómico 16S/genética
7.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L1035-L1048, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30838865

RESUMEN

Protein phosphatase 2A (PP2A), a ubiquitously expressed Ser/Thr phosphatase is an important regulator of cytokine signaling and cell function. We previously showed that myeloid-specific deletion of PP2A (LysMcrePP2A-/-) increased mortality in a murine peritoneal sepsis model. In the current study, we assessed the role of myeloid PP2A in regulation of lung injury induced by lipopolysaccharide (LPS) or bleomycin delivered intratracheally. LysMcrePP2A-/- mice experienced increased lung injury in response to both LPS and bleomycin. LysMcrePP2A-/- mice developed more exuberant fibrosis in response to bleomycin, elevated cytokine responses, and chronic myeloid inflammation. Bone marrow-derived macrophages (BMDMs) from LysMcrePP2A-/- mice showed exaggerated inflammatory cytokine release under conditions of both M1 and M2 activation. Notably, secretion of IL-10 was elevated under all stimulation conditions, including activation of BMDMs by multiple Toll-like receptor ligands. Supernatants collected from LPS-stimulated LysMcrePP2A-/- BMDMs induced epithelial cell apoptosis in vitro but this effect was mitigated when IL-10 was also depleted from the BMDMs by crossing LysMcrePP2A-/- mice with systemic IL-10-/- mice (LysMcrePP2A-/- × IL-10-/-) or when IL-10 was neutralized. Despite these findings, IL-10 did not directly induce epithelial cell apoptosis but sensitized epithelial cells to other mediators from the BMDMs. Taken together our results demonstrate that myeloid PP2A regulates production of multiple cytokines but that its effect is most pronounced on IL-10 production. Furthermore, IL-10 sensitizes epithelial cells to apoptosis in response to myeloid-derived mediators, which likely contributes to the pathogenesis of lung injury and fibrosis in this model.


Asunto(s)
Células Epiteliales/metabolismo , Interleucina-10/metabolismo , Lesión Pulmonar/patología , Proteína Fosfatasa 2/genética , Fibrosis Pulmonar/patología , Animales , Apoptosis/genética , Bleomicina/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Lipopolisacáridos/toxicidad , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/genética , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Síndrome de Dificultad Respiratoria/patología
8.
J Virol ; 92(23)2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30232191

RESUMEN

Noroviruses are enteric pathogens causing significant morbidity, mortality, and economic losses worldwide. Secretory immunoglobulins (sIg) are a first line of mucosal defense against enteric pathogens. They are secreted into the intestinal lumen via the polymeric immunoglobulin receptor (pIgR), where they bind to antigens. However, whether natural sIg protect against norovirus infection remains unknown. To determine if natural sIg alter murine norovirus (MNV) pathogenesis, we infected pIgR knockout (KO) mice, which lack sIg in mucosal secretions. Acute MNV infection was significantly reduced in pIgR KO mice compared to controls, despite increased MNV target cells in the Peyer's patch. Natural sIg did not alter MNV binding to the follicle-associated epithelium (FAE) or crossing of the FAE into the lymphoid follicle. Instead, naive pIgR KO mice had enhanced levels of the antiviral inflammatory molecules interferon gamma (IFN-γ) and inducible nitric oxide synthase (iNOS) in the ileum compared to controls. Strikingly, depletion of the intestinal microbiota in pIgR KO and control mice resulted in comparable IFN-γ and iNOS levels, as well as MNV infectious titers. IFN-γ treatment of wild-type (WT) mice and neutralization of IFN-γ in pIgR KO mice modulated MNV titers, implicating the antiviral cytokine in the phenotype. Reduced gastrointestinal infection in pIgR KO mice was also observed with another enteric virus, reovirus. Collectively, our findings suggest that natural sIg are not protective during enteric virus infection, but rather, that sIg promote enteric viral infection through alterations in microbial immune responses.IMPORTANCE Enteric virus, such as norovirus, infections cause significant morbidity and mortality worldwide. However, direct antiviral infection prevention strategies are limited. Blocking host entry and initiation of infection provides an established avenue for intervention. Here, we investigated the role of the polymeric immunoglobulin receptor (pIgR)-secretory immunoglobulin (sIg) cycle during enteric virus infections. The innate immune functions of sIg (agglutination, immune exclusion, neutralization, and expulsion) were not required during control of acute murine norovirus (MNV) infection. Instead, lack of pIgR resulted in increased IFN-γ levels, which contributed to reduced MNV titers. Another enteric virus, reovirus, also showed decreased infection in pIgR KO mice. Collectively, our data point to a model in which sIg-mediated microbial sensing promotes norovirus and reovirus infection. These data provide the first evidence of the proviral role of natural sIg during enteric virus infections and provide another example of how intestinal bacterial communities indirectly influence MNV pathogenesis.


Asunto(s)
Infecciones por Caliciviridae/virología , Tracto Gastrointestinal/virología , Inmunoglobulinas/metabolismo , Receptores de Inmunoglobulina Polimérica/fisiología , Infecciones por Reoviridae/virología , Replicación Viral/inmunología , Animales , Infecciones por Caliciviridae/inmunología , Infecciones por Caliciviridae/metabolismo , Tracto Gastrointestinal/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Norovirus/inmunología , Reoviridae/inmunología , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/metabolismo
9.
Am J Respir Crit Care Med ; 198(10): 1312-1321, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29878854

RESUMEN

RATIONALE: Hematopoietic cell transplant (HCT) is a common treatment for hematological neoplasms and autoimmune disorders. Among HCT recipients, pulmonary complications are common, morbid, and/or lethal, and they have recently been associated with gut dysbiosis. The role of lung microbiota in post-HCT pulmonary complications is unknown. OBJECTIVES: To investigate the role of lung microbiota in post-HCT pulmonary complications using animal modeling and human BAL fluid. METHODS: For animal modeling, we used an established murine model of HCT with and without postengraftment herpes virus infection. For human studies, we characterized lung microbiota in BAL fluid from 43 HCT recipients. Lung bacteria were characterized using 16S ribosomal RNA gene sequencing and were compared with lung histology (murine) and with alveolar inflammation and pulmonary function testing (human). MEASUREMENTS AND MAIN RESULTS: Both HCT and viral infection independently altered the composition of murine lung microbiota, but they had no effect on lung microbial diversity. By contrast, combined HCT and viral infection profoundly altered lung microbiota, decreasing community diversity with an associated pneumonitis. Among human HCT recipients, increased relative abundance of the Proteobacteria phylum was associated with impaired pulmonary function, and lung microbiota were significantly associated with alveolar concentrations of inflammatory cytokines. CONCLUSIONS: In animal models and human subjects, lung dysbiosis is a prominent feature of HCT. Lung dysbiosis is correlated with histologic, immunologic, and physiologic features of post-HCT pulmonary complications. Our findings suggest the lung microbiome may be an unappreciated target for the prevention and treatment of post-HCT pulmonary complications.


Asunto(s)
Disbiosis/epidemiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Inflamación/epidemiología , Enfermedades Pulmonares/epidemiología , Complicaciones Posoperatorias/epidemiología , Animales , Comorbilidad , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal , Humanos , Inflamación/microbiología , Pulmón/microbiología , Enfermedades Pulmonares/microbiología , Masculino , Ratones , Persona de Mediana Edad , Complicaciones Posoperatorias/microbiología
10.
FASEB J ; 31(12): 5543-5556, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28821630

RESUMEN

Interleukin 17A (IL-17A) and complement (C') activation have each been implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF). We have reported that IL-17A induces epithelial injury via TGF-ß in murine bronchiolitis obliterans; that TGF-ß and the C' cascade present signaling interactions in mediating epithelial injury; and that the blockade of C' receptors mitigates lung fibrosis. In the present study, we investigated the role of IL-17A in regulating C' in lung fibrosis. Microarray analyses of mRNA isolated from primary normal human small airway epithelial cells indicated that IL-17A (100 ng/ml; 24 h; n = 5 donor lungs) induces C' components (C' factor B, C3, and GPCR kinase isoform 5), cytokines (IL8, -6, and -1B), and cytokine ligands (CXCL1, -2, -3, -5, -6, and -16). IL-17A induces protein and mRNA regulation of C' components and the synthesis of active C' 3a (C3a) in normal primary human alveolar type II epithelial cells (AECs). Wild-type mice subjected to IL-17A neutralization and IL-17A knockout (il17a-/- ) mice were protected against bleomycin (BLEO)-induced fibrosis and collagen deposition. Further, BLEO-injured il17a-/- mice had diminished levels of circulating Krebs Von Den Lungen 6 (alveolar epithelial injury marker), local caspase-3/7, and local endoplasmic reticular stress-related genes. BLEO-induced local C' activation [C3a, C5a, and terminal C' complex (C5b-9)] was attenuated in il17a-/- mice, and IL-17A neutralization prevented the loss of epithelial C' inhibitors (C' receptor-1 related isoform Y and decay accelerating factor), and an increase in local TUNEL levels. RNAi-mediated gene silencing of il17a in fibrotic mice arrested the progression of lung fibrosis, attenuated cellular apoptosis (caspase-3/7) and lung deposition of collagen and C' (C5b-9). Compared to normals, plasma from IPF patients showed significantly higher hemolytic activity. Our findings demonstrate that limiting complement activation by neutralizing IL-17A is a potential mechanism in ameliorating lung fibrosis.-Cipolla, E., Fisher, A. J., Gu, H., Mickler, E. A., Agarwal, M., Wilke, C. A., Kim, K. K., Moore, B. B., Vittal, R. IL-17A deficiency mitigates bleomycin-induced complement activation during lung fibrosis.


Asunto(s)
Bleomicina/farmacología , Activación de Complemento/efectos de los fármacos , Fibrosis/metabolismo , Interleucina-17/deficiencia , Interleucina-17/metabolismo , Enfermedades Pulmonares/metabolismo , Anciano , Animales , Western Blotting , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrosis/genética , Técnica del Anticuerpo Fluorescente , Hemólisis/genética , Hemólisis/fisiología , Humanos , Interleucina-17/genética , Enfermedades Pulmonares/genética , Masculino , Ratones , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Am J Respir Cell Mol Biol ; 54(4): 482-92, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26378893

RESUMEN

Accumulation of apoptosis-resistant fibroblasts is a hallmark of pulmonary fibrosis. We hypothesized that disruption of inhibitor of apoptosis protein (IAP) family proteins would limit lung fibrosis. We first show that transforming growth factor-ß1 and bleomycin increase X-linked IAP (XIAP) and cellular IAP (cIAP)-1 and -2 in murine lungs and mesenchymal cells. Functional blockade of XIAP and the cIAPs with AT-406, an orally bioavailable second mitochondria-derived activator of caspases (Smac) mimetic, abrogated bleomycin-induced lung fibrosis when given both prophylactically and therapeutically. To determine whether the reduction in fibrosis was predominantly due to AT-406-mediated inhibition of XIAP, we compared the fibrotic response of XIAP-deficient mice (XIAP(-/y)) with littermate controls and found no difference. We found no alterations in total inflammatory cells of either wild-type mice treated with AT-406 or XIAP(-/y) mice. AT-406 treatment limited CCL12 and IFN-γ production, whereas XIAP(-/y) mice exhibited increased IL-1ß expression. Surprisingly, XIAP(-/y) mesenchymal cells had increased resistance to Fas-mediated apoptosis. Functional blockade of cIAPs with AT-406 restored sensitivity to Fas-mediated apoptosis in XIAP(-/y) mesenchymal cells in vitro and increased apoptosis of mesenchymal cells in vivo, indicating that the increased apoptosis resistance in XIAP(-/y) mesenchymal cells was the result of increased cIAP expression. Collectively, these results indicate that: (1) IAPs have a role in the pathogenesis of lung fibrosis; (2) a congenital deficiency of XIAP may be overcome by compensatory mechanisms of other IAPs; and (3) broad functional inhibition of IAPs may be an effective strategy for the treatment of lung fibrosis by promoting mesenchymal cell apoptosis.


Asunto(s)
Bleomicina/toxicidad , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Fibrosis Pulmonar/prevención & control , Animales , Apoptosis , Azocinas/farmacología , Compuestos de Bencidrilo/farmacología , Proteínas Inhibidoras de la Apoptosis/genética , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quimioatrayentes de Monocitos/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta1/administración & dosificación
12.
Am J Physiol Lung Cell Mol Physiol ; 311(3): L611-27, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27448666

RESUMEN

CCR2-expressing leukocytes are required for the progression of fibrosis in models of induced lung injury as well as models of bone marrow transplant (BMT)-related idiopathic pneumonia syndrome. Infection with murid γ-herpesvirus-68 (γHV-68) results in severe pneumonitis and pulmonary fibrosis following syngeneic BMT; however, the roles that various proinflammatory leukocyte populations play in this process remain unclear. Deletion of CCR2 in both non-BMT and BMT mice increased early lytic viral replication and resulted in a reduction in the numbers of lung-infiltrating GR1+,F4/80+ and CXCR1+ cells, while maintaining robust neutrophil infiltration. Similarly, in γHV-68-infected CCR2(-/-) BMT mice, recruitment of monocytes and lymphocytes were reduced whereas neutrophil recruitment was increased compared with wild-type (WT) BMT mice. Interestingly, levels of profibrotic IL-17 were increased in infected CCR2 BMT mice compared with WT BMT. Furthermore, an increase in lung-associated collagen was detected even though there was an overall decrease in the number of profibrotic CCR2+ fibrocytes detected in the lungs of CCR2(-/-) BMT mice. These data indicate that, contrary to most models of fibrosis, deletion of CCR2 offers no protection from γ-herpesvirus-induced pneumonitis and fibrosis, and, indeed, CCR2+ cells play a suppressive role during the development of pulmonary fibrosis following γ-herpesvirus infection post-BMT by limiting IL-7 and collagen production.


Asunto(s)
Infecciones por Herpesviridae/metabolismo , Neumonía Viral/metabolismo , Fibrosis Pulmonar/metabolismo , Receptores CCR2/fisiología , Animales , Trasplante de Médula Ósea , Células Cultivadas , Gammaherpesvirinae/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Neumonía Viral/inmunología , Neumonía Viral/virología , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/virología , Transducción de Señal , Replicación Viral
13.
J Immunol ; 193(8): 4245-53, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25225663

RESUMEN

The role and origin of alveolar macrophages (AMs) in asthma are incompletely defined. We sought to clarify these issues in the context of acute allergic lung inflammation using house dust mite and OVA murine models. Use of liposomal clodronate to deplete resident AMs (rAMs) resulted in increased levels of inflammatory cytokines and eosinophil numbers in lavage fluid and augmented the histopathologic evidence of lung inflammation, suggesting a suppressive role for rAMs. Lung digests of asthmatic mice revealed an increased percentage of Ly6C(high)/CD11b(pos) inflammatory monocytes. Clodronate depletion of circulating monocytes, by contrast, resulted in an attenuation of allergic inflammation. A CD45.1/CD45.2 chimera model demonstrated that recruitment at least partially contributes to the AM pool in irradiated nonasthmatic mice, but its contribution was no greater in asthma. Ki-67 staining of AMs supported a role for local proliferation, which was increased in asthma. Our data demonstrate that rAMs dampen, whereas circulating monocytes promote, early events in allergic lung inflammation. Moreover, maintenance of the AM pool in the early stages of asthmatic inflammation depends on local proliferation, but not recruitment.


Asunto(s)
Asma/inmunología , Inflamación/inmunología , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Alérgenos/inmunología , Alveolitis Alérgica Extrínseca/inmunología , Animales , Antígenos Ly/biosíntesis , Líquido del Lavado Bronquioalveolar/citología , Antígeno CD11b/biosíntesis , Proliferación Celular , Ácido Clodrónico/farmacología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Antígenos Comunes de Leucocito/genética , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Neumonía/inmunología , Pyroglyphidae/inmunología
14.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L86-95, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25361568

RESUMEN

Hematopoietic stem cell transplantation (HSCT) is complicated by pulmonary infections that manifest posttransplantation. Despite engraftment, susceptibility to infections persists long after reconstitution. Previous work using a murine bone marrow transplant (BMT) model implicated increased cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) in promoting impaired alveolar macrophage (AM) responses. However, mechanisms driving COX-2 overexpression remained elusive. Previously, transforming growth factor-ß (TGF-ß) signaling after BMT was shown to promote hypomethylation of the COX-2 gene. Here, we provide mechanistic insight into how this occurs and show that TGF-ß induces microRNA (miR)-29b while decreasing DNA methyltransferases (DNMT)1, DNMT3a, and DNMT3b in AMs after BMT. De novo DNMT3a and DNMT3b were decreased upon transient transfection of miR-29b, resulting in decreased methylation of the COX-2 promoter and induction of COX-2. As a consequence, miR-29b-driven upregulation of COX-2 promoted AM dysfunction, and transfection of BMT AMs with a miR-29b inhibitor rescued the bacterial-killing defect. MiR-29b-mediated defects in BMT AMs were dependent on increased levels of PGE2, as miR-29b-transfected AMs treated with a novel E prostanoid receptor 2 antagonist abrogated the impaired bacterial killing. We also demonstrate that patients that have undergone HSCT exhibit increased miR-29b; thus these studies highlight miR-29b in driving defective AM responses and identify this miRNA as a potential therapeutic target.


Asunto(s)
Trasplante de Médula Ósea , Trasplante de Células Madre Hematopoyéticas , Macrófagos Alveolares/metabolismo , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Aloinjertos , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Dinoprostona/genética , Dinoprostona/metabolismo , Femenino , Humanos , Macrófagos Alveolares/patología , Masculino , Ratones , MicroARNs/genética , Persona de Mediana Edad , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética
15.
J Immunol ; 189(9): 4528-36, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23008450

RESUMEN

Hematopoietic stem cell transplant therapy is limited by pulmonary infections. Mice with fully reconstituted hematopoietic compartments, including alveolar macrophages (AMs), after bone marrow transplantation (BMT) have impaired host defense against Gram-negative Pseudomonas aeruginosa. Impaired innate immunity is related to increased production of PGE(2) by AMs. Cyclooxygenase (COX)-2 is the rate-limiting enzyme for synthesis of PGE(2) from arachidonic acid, and COX-2 expression is elevated in AMs post-BMT. We hypothesized that epigenetic mechanisms may be responsible for upregulation of COX-2 in AMs. Using bisulfite sequencing, we observed the 5'-untranslated region and exon 1 of the COX-2 gene is hypomethylated in the AMs of BMT mice compared with control. COX-2 expression was increased in primary AMs and in the AM cell line (MHS) after treatment with 5-aza-2'-deoxycytidine (a methyltransferase inhibitor). Methylation by SssI methyltransferase of a 698-bp region of the COX-2 promoter including the beginning of exon 1 driving a luciferase reporter silenced luciferase expression. Because TGF-ß1 is elevated in lungs post-BMT, we tested whether TGF-ß1 could promote expression of COX-2 in a hypermethylated COX-2 vector, and observed TGF-ß1-induced modest expression of COX-2, suggesting an ability to demethylate the promoter. Finally, BMTs performed with marrow from mice expressing a dominant-negative form of the TGF-ßRII on CD11c-expressing cells (which includes AMs) demonstrated improved host defense and AM function. Our findings suggest impaired innate immunity and PGE(2) elevation post-BMT are due to hypomethylation of the COX-2 gene, which is at least partly regulated by TGF-ß1.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Metilación de ADN/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Regiones no Traducidas 5'/inmunología , Animales , Línea Celular , Ciclooxigenasa 2/deficiencia , Metilación de ADN/genética , Exones/inmunología , Técnicas de Silenciamiento del Gen , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología
16.
Am J Physiol Lung Cell Mol Physiol ; 303(12): L1046-56, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23043074

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease without effective therapeutics. Periostin has been reported to be elevated in IPF patients relative to controls, but its sources and mechanisms of action remain unclear. We confirm excess periostin in lungs of IPF patients and show that IPF fibroblasts produce periostin. Blood was obtained from 54 IPF patients (all but 1 with 48 wk of follow-up). We show that periostin levels predict clinical progression at 48 wk (hazard ratio = 1.47, 95% confidence interval = 1.03-2.10, P < 0.05). Monocytes and fibrocytes are sources of periostin in circulation in IPF patients. Previous studies suggest that periostin may regulate the inflammatory phase of bleomycin-induced lung injury, but periostin effects during the fibroproliferative phase of the disease are unknown. Wild-type and periostin-deficient (periostin(-/-)) mice were anesthetized and challenged with bleomycin. Wild-type mice were injected with bleomycin and then treated with OC-20 Ab (which blocks periostin and integrin interactions) or control Ab during the fibroproliferative phase of disease, and fibrosis and survival were assessed. Periostin expression was upregulated quickly after treatment with bleomycin and remained elevated. Periostin(-/-) mice were protected from bleomycin-induced fibrosis. Instillation of OC-20 during the fibroproliferative phase improved survival and limited collagen deposition. Chimeric mouse studies suggest that hematopoietic and structural sources of periostin contribute to lung fibrogenesis. Periostin was upregulated by transforming growth factor-ß in lung mesenchymal cells, and periostin promoted extracellular matrix deposition, mesenchymal cell proliferation, and wound closure. Thus periostin plays a vital role in late stages of pulmonary fibrosis and is a potential biomarker for disease progression and a target for therapeutic intervention.


Asunto(s)
Moléculas de Adhesión Celular/sangre , Fibrosis Pulmonar Idiopática/metabolismo , Anciano , Animales , Anticuerpos Neutralizantes/farmacología , Biomarcadores , Moléculas de Adhesión Celular/biosíntesis , Proliferación Celular , Colágeno/metabolismo , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Monocitos/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Cicatrización de Heridas
17.
Am J Pathol ; 179(5): 2382-96, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21924228

RESUMEN

Pulmonary infections and pneumonitis occur frequently after hematopoietic stem cell transplantation. Using a syngeneic mouse model of bone marrow transplantation (BMT), we have previously demonstrated that BMT mice are more susceptible to acute gammaherpesvirus 68 (MHV-68) replication at day 7 after infection. By day 21, the virus is latent in lungs of BMT and control mice, and there is no difference in viral load. Despite similar latent viral load, BMT mice develop severe pneumonitis associated with reduced oxygen saturation, fibrosis, peripheral inflammation, hyaline membranes, and foamy alveolar macrophages, a phenotype that persists for 7 weeks after infection. BMT mice demonstrate increased bronchoalveolar lavage (BAL) cells, and this population is enriched in neutrophils and T cells. Alternatively, activated macrophages appear earlier than do classically activated macrophages. BAL fluid from BMT mice at day 21 after infection contains increased levels of hydrogen peroxide, nitrite, and transforming growth factor-ß (TGF-ß). Mice expressing the dominant-negative transgene dn-TGFßRII in multiple cell types were used as BMT donors. BMT mice with T-cell dnTGFßRII are largely protected from the pneumonitis phenotype, whereas mice with CD11c-dnTGFßRII BMT mice are only modestly protected from pneumonitis. Protection in BMT mice with T-cell dnTGFßRII is associated with decreased TGF-ß derived from parenchymal cells in the BAL fluid, lower nitrite levels, and reduced apoptosis, whereas alternatively activated macrophage markers are unchanged.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Gammaherpesvirinae , Infecciones por Herpesviridae , Neumonía/virología , Fibrosis Pulmonar/virología , Factor de Crecimiento Transformador beta/fisiología , Animales , Apoptosis/fisiología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/virología , Activación de Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Transducción de Señal/fisiología , Linfocitos T Reguladores/fisiología , Transgenes , Carga Viral
18.
J Immunol ; 184(9): 5130-40, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20348421

RESUMEN

Patients receiving hematopoietic stem cell transplantation or bone marrow transplantation (BMT) as therapy for various malignancies or autoimmune diseases have an increased risk for infectious complications posttransplant, especially in the lung. We have used BMT in mice and murine gammaherpesvirus, gammaHV-68, to study the efficacy of adaptive immune responses post-BMT. Five weeks posttransplant, mice have fully reconstituted their hematopoietic lineages in both the lung and periphery. When challenged with virus, however, BMT mice have a reduced ability to clear lytic virus from the lung. Defective viral control in BMT mice is not related to impaired leukocyte recruitment or defective APC function. Rather, BMT mice are characterized by defective CD4 cell proliferation, skewing of effector CD4 T cells from a Th1 to a Th17 phenotype, and an immunosuppressive lung environment at the time of infection that includes overexpression of TGF-beta1 and PGE(2) and increased numbers of regulatory T cells. Neither indomethacin treatment to block PG synthesis nor anti-CD25 depletion of regulatory T cells improved antiviral host defense post-BMT. Transplanting mice with transgenic bone marrow expressing a dominant-negative TGF-betaRII under the permissive CD4 promoter created mice in which effector CD4 and CD8 cells were unresponsive to TGF-beta1. Mice with TGF-beta1-nonresponsive effector T cells had restored antiviral immunity and improved Th1 responses post-BMT. Thus, our results indicate that overexpression of TGF-beta1 following myeloablative conditioning post-BMT results in impaired effector T cell responses to viral infection.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Infecciones por Herpesviridae/inmunología , Neumonía Viral/inmunología , Rhadinovirus/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/efectos adversos , Factor de Crecimiento Transformador beta1/biosíntesis , Infecciones Tumorales por Virus/inmunología , Animales , Trasplante de Médula Ósea/efectos adversos , Diferenciación Celular/inmunología , Proliferación Celular , Inhibidores de Crecimiento/efectos adversos , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/genética , Infecciones por Herpesviridae/terapia , Infecciones por Herpesviridae/virología , Interleucina-17/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Neumonía Viral/terapia , Neumonía Viral/virología , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/uso terapéutico , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/administración & dosificación , Receptores de Factores de Crecimiento Transformadores beta/uso terapéutico , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/virología , Células TH1/inmunología , Células TH1/patología , Células TH1/virología , Factor de Crecimiento Transformador beta1/genética , Acondicionamiento Pretrasplante/métodos , Infecciones Tumorales por Virus/terapia , Infecciones Tumorales por Virus/virología
19.
J Immunol ; 184(11): 6299-308, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20439918

RESUMEN

Following immune reconstitution, hematopoietic stem cell transplant patients often display reduced immune function and are especially susceptible to lung infections. In a mouse model of syngeneic bone marrow transplantation (BMT), we previously reported that PGE(2) is overproduced in lungs of BMT mice, significantly impairing host defense against Pseudomonas aeruginosa. This impairment in host defense post-BMT is also marked by diminished alveolar macrophage (AM) phagocytosis, bacterial killing, and production of TNF-alpha and cysteinyl leukotrienes. However, a mechanism by which overproduction of PGE(2) suppresses pulmonary host defense post-BMT is unknown. As IL-1R-associated kinase (IRAK)-M is a known inhibitor of MyD88-dependent IL-1R/TLR signaling and macrophage function, we sought to determine whether IRAK-M is involved in PGE(2)-induced immunosuppression post-BMT. We found that IRAK-M expression is elevated 3.5-fold in BMT AMs relative to control AMs, and this is related to AM overproduction of PGE(2). Furthermore, genetic ablation of IRAK-M in the bone marrow of BMT mice restores host defense against P. aeruginosa. Despite AM overproduction of PGE(2) and elevated E prostanoid 2 receptor expression, AM phagocytosis, killing, and production of cysteinyl leukotrienes and TNF-alpha are restored in the absence of IRAK-M post-BMT. Also, treatment with PGE(2) does not inhibit AM phagocytosis in the absence of IRAK-M. These data suggest that the absence of IRAK-M in the hematopoietic compartment post-BMT enhances pulmonary host defense and mitigates AM sensitivity to the inhibitory effects of PGE(2). Therefore, strategies to limit IRAK-M elevation post-BMT may be efficacious in reducing patient susceptibility to infection.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Dinoprostona/inmunología , Tolerancia Inmunológica/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Macrófagos Alveolares/inmunología , Animales , Separación Celular , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Am J Respir Cell Mol Biol ; 45(5): 1050-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21527775

RESUMEN

Hematopoietic stem cell transplant patients are susceptible to infection despite cellular reconstitution. In a murine model of syngeneic bone marrow transplantation (BMT), we previously reported that BMT mice have impaired host defense against Pseudomonas aeruginosa pneumonia due to overproduction of (PG)E(2) in lung. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is an effector in the PGE(2) signaling pathway that negatively regulates alveolar macrophage (AM) phagocytosis and bacterial killing. Therefore, examined whether overproduction of PGE(2) after BMT inhibits AM host defense by up-regulating PTEN phosphatase activity. We found that PTEN activity is elevated in BMT AMs in response to increased PGE(2) signaling and that pharmacological inhibition of PTEN activity in BMT AMs fully restores phagocytosis of serum-opsonized P. aeruginosa but only partially restores phagocytosis of nonopsonized P. aeruginosa. In wild-type mice transplanted with myeloid-specific conditional PTEN knockout (PTEN CKO) bone marrow, bacterial clearance is improved after challenge with P. aeruginosa pneumonia. Furthermore, PTEN CKO BMT AMs display improved TNF-α production and enhanced phagocytosis and killing of serum-opsonized P. aeruginosa despite overproduction of PGE(2). However, AM phagocytosis of nonopsonized P. aeruginosa is only partially restored in the absence of PTEN after BMT. This may be related to elevated AM expression of IL-1 receptor-associated kinase (IRAK)-M, a molecule previously identified in the PGE(2) signaling pathway to inhibit AM phagocytosis of nonopsonized bacteria. These data suggest that PGE(2) signaling up-regulates IRAK-M independently of PTEN and that these molecules differentially inhibit opsonized and nonopsonized phagocytosis of P. aeruginosa.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Macrófagos Alveolares/inmunología , Fosfohidrolasa PTEN/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Animales , Antiinflamatorios no Esteroideos/farmacología , Dinoprostona/inmunología , Inhibidores Enzimáticos/farmacología , Indometacina/farmacología , Quinasas Asociadas a Receptores de Interleucina-1/biosíntesis , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Compuestos Organometálicos/farmacología , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/metabolismo , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/inmunología , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA