Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Front Immunol ; 14: 1094464, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36845149

RESUMO

ß-glucan is the most abundant polysaccharide in the cell wall of Pneumocystis jirovecii, which has attracted extensive attention because of its unique immunobiological characteristics. ß-glucan binds to various cell surface receptors, which produces an inflammatory response and accounts for its immune effects. A deeper comprehension of the processes by Pneumocystis ß-glucan recognizes its receptors, activates related signaling pathways, and regulates immunity as required. Such understanding will provide a basis for developing new therapies against Pneumocystis. Herein, we briefly review the structural composition of ß-glucans as a vital component of the Pneumocystis cell wall, the host immunity mediated by ß-glucans after their recognition, and discuss opportunities for the development of new strategies to combat Pneumocystis.


Assuntos
Pneumocystis , Pneumonia por Pneumocystis , beta-Glucanas , Glucanos , Pneumonia por Pneumocystis/metabolismo , Parede Celular
2.
Front Immunol ; 12: 798214, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975910

RESUMO

Pneumocystis jirovecii is one of the most common fungal pathogens in immunocompromised individuals. Pneumocystis jirovecii pneumonia (PJP) causes a significant host immune response that is driven greatly by the organism's cell wall components including ß-glucans and major surface glycoprotein (Msg). These ligands interact with a number of C-type lectin receptors (CLRs) leading to downstream activation of proinflammatory signaling pathways. This minireview provides a brief overview summarizing known CLR/Pneumocystis interactions.


Assuntos
Proteínas Fúngicas/imunologia , Imunidade Inata , Lectinas Tipo C/imunologia , Glicoproteínas de Membrana/imunologia , Pneumocystis carinii/imunologia , Pneumonia por Pneumocystis/imunologia , beta-Glucanas/imunologia , Animais , Proteínas Fúngicas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Lectinas Tipo C/metabolismo , Ligantes , Glicoproteínas de Membrana/metabolismo , Pneumocystis carinii/metabolismo , Pneumocystis carinii/patogenicidade , Pneumonia por Pneumocystis/metabolismo , Pneumonia por Pneumocystis/microbiologia , Transdução de Sinais , beta-Glucanas/metabolismo
4.
Cell Microbiol ; 22(10): e13235, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32548948

RESUMO

Caspase recruitment domains-containing protein 9 (CARD9) is an adaptor molecule critical for key signalling pathways initiated through C-type lectin receptors (CLRs). Previous studies demonstrated that Pneumocystis organisms are recognised through a variety of CLRs. However, the role of the downstream CARD9 adaptor signalling protein in host defence against Pneumocystis infection remains to be elucidated. Herein, we analysed the role of CARD9 in host defence against Pneumocystis both in CD4-depleted CARD9-/- and immunocompetent hosts. Card9 gene-disrupted (CARD9-/- ) mice were more susceptible to Pneumocystis, as evidenced by reduced fungal clearance in infected lungs compared to wild-type (WT) infected mice. Our data suggests that this defect was due to impaired proinflammatory responses. Furthermore, CARD9-/- macrophages were severely compromised in their ability to differentiate and express M1 and M2 macrophage polarisation markers, to enhanced mRNA expression for Dectin-1 and Mincle, and most importantly, to kill Pneumocystis in vitro. Remarkably, compared to WT mice, and despite markedly increased organism burdens, CARD9-/- animals did not exhibit worsened survival during pneumocystis pneumonia (PCP), perhaps related to decreased lung injury due to altered influx of inflammatory cells and decreased levels of proinflammatory cytokines in response to the organism. Finally, although innate phase cytokines were impaired in the CARD9-/- animals during PCP, T-helper cell cytokines were normal in immunocompetent CARD9-/- animals infected with Pneumocystis. Taken together, our data demonstrate that CARD9 has a critical function in innate immune responses against Pneumocystis.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Macrófagos Alveolares/imunologia , Pneumocystis carinii/imunologia , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Diferenciação Celular , Contagem de Colônia Microbiana , Citocinas/metabolismo , Hospedeiro Imunocomprometido , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Pulmão/enzimologia , Pulmão/microbiologia , Pulmão/patologia , Macrófagos Alveolares/citologia , Macrófagos Alveolares/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Peroxidase/metabolismo , Pneumocystis/crescimento & desenvolvimento , Pneumocystis carinii/crescimento & desenvolvimento , Pneumonia por Pneumocystis/microbiologia , Pneumonia por Pneumocystis/patologia , Ratos , Linfócitos T Auxiliares-Indutores/imunologia , Fator de Necrose Tumoral alfa/metabolismo
6.
Sci Rep ; 9(1): 2078, 2019 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-30765827

RESUMO

Airway mucus responses to subclinical infections may explain variations in progression of chronic lung diseases and differences in clinical expression of respiratory infections across individuals. Pneumocystis associates to more severe Chronic Obstructive Pulmonary Disease (COPD), asthma, respiratory distress of premature newborns, and is a consistent subclinical infection between 2 and 5 months of age when hospitalizations for respiratory cause and infant mortality are higher. This atypical fungus associates to increased mucin 5AC (MUC5AC), a central effector of Th2-type allergic inflammation, in infant lungs. However, mucus progression, expression of MUC5B essential for airway defense, and potential for pharmacologic modulation of mucus during Pneumocystis infection remain unknown. We measured MUC5B and Pneumocystis in infant lungs, and progression of mucin levels and effect of inhibition of the STAT6/FoxA2 mucus pathway using Kaempferol, a JAK/STAT6 inhibitor, in immunocompetent rats during Pneumocystis primary infection. Pneumocystis associated to increased MUC5B in infant lungs. Muc5b increased earlier and more abundantly than Muc5ac during experimental primary infection suggesting an acute defensive response against Pneumocystis as described against bacteria, while increased Muc5ac levels supports an ongoing allergic, Th2 lymphocyte-type response during primary Pneumocystis infection. Kaempferol partly reversed Muc5b stimulation suggesting limited potential for pharmacological modulation via the STAT6-FoxA2 pathway.


Assuntos
Mucina-5B/metabolismo , Infecções por Pneumocystis/metabolismo , Mucosa Respiratória/metabolismo , Animais , Asma/metabolismo , Células Epiteliais/metabolismo , Feminino , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Lactente , Recém-Nascido , Inflamação/metabolismo , Pulmão/metabolismo , Masculino , Mucina-5B/genética , Mucinas/genética , Mucinas/metabolismo , Muco/metabolismo , Pneumocystis/patogenicidade , Pneumonia por Pneumocystis/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT6/metabolismo
7.
BMC Pulm Med ; 19(1): 47, 2019 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-30791907

RESUMO

BACKGROUND: In patients with non-HIV Pneumocystis jirovecii pneumonia (PjP), computed tomography imaging reveals ground grass opacities (GGO). Previous reports show that some patients with non-HIV PjP exhibit GGO with crazy paving. However, there have been no studies on the association between crazy paving GGO and non-HIV PjP clinical outcomes. Here, at the diagnosis of non-HIV PjP, we reviewed high-resolution computed tomography (HRCT) findings that included GGO types and evaluated the prognostic impact of crazy paving GGO on the clinical outcomes of non-HIV PjP immunocompromised patients. METHODS: We retrospectively reviewed the clinical information including the HRCT findings of patients diagnosed with non-HIV PjP from five institutions between 2006 and 2015. The GGO types included those with or without crazy paving. The associations between clinical factors such as HRCT findings and in-hospital mortality were assessed using the Cox regression model. RESULTS: Sixty-one patients were included in our study. Nineteen patients died at a hospital. All patients exhibited GGO on HRCT imaging at diagnosis of non-HIV PjP. The HRCT findings included crazy paving GGO (29 patients, 47.5%), consolidations (23 patients, 37.7%), bronchiectasis (14 patients, 23.0%), and centrilobular small nodules (30 patients, 49.2%). Cysts were not observed in any patient. Multivariate analysis revealed that crazy paving GGO and low serum albumin levels were independent risk factors for mortality. CONCLUSIONS: At the diagnosis of non-HIV PjP, patients with crazy paving GGO on HRCT imaging and low serum albumin levels may have a poor prognosis.


Assuntos
Mortalidade Hospitalar , Pulmão/diagnóstico por imagem , Pneumonia por Pneumocystis/diagnóstico por imagem , Corticosteroides/efeitos adversos , Idoso , Antineoplásicos/efeitos adversos , Doenças Autoimunes/imunologia , Estudos de Coortes , Doenças do Tecido Conjuntivo/imunologia , Feminino , Humanos , Hospedeiro Imunocomprometido , Imunossupressores/efeitos adversos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Neoplasias/imunologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/metabolismo , Pneumonia por Pneumocystis/mortalidade , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Albumina Sérica/metabolismo , Tomografia Computadorizada por Raios X
9.
Sci Rep ; 8(1): 8321, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29844519

RESUMO

Rituximab is associated with a higher incidence of Pneumocystis jirovecii pneumonia infection. Pneumocystis prophylaxis is advised in many immunocompromised populations treated with rituximab. However, the beneficial effect of pneumocystis prophylaxis in HIV-uninfected, rituximab-treated non-Hodgkin lymphoma (NHL) patients has not been assessed. Thus, we conducted this retrospective study to explore pneumocystis infection in HIV-uninfected NHL patients who received at least three courses of chemotherapy without haematopoietic stem cell transplantation using the Taiwan National Health Insurance Research Database. Patients who had rituximab-based chemotherapy were included in the experimental (rituximab) group, while the rest of the patients who did not receive any rituximab-based chemotherapy throughout the study period formed the control group. The prevalence rate of pneumocystis infection in the rituximab group (N = 7,554) was significantly higher than that in the control group (N = 4,604) (2.95% vs. 1.32%). The onset of pneumocystis infection occurred between 6 and 16 weeks after chemotherapy. Patients who had pneumocystis prophylaxis, whether or not they had a pneumocystis infection later in their treatment course, had significantly better first-year survival rates (73% vs. 38%). Regular pneumocystis prophylaxis should be considered in this group of patients.


Assuntos
Pneumonia por Pneumocystis/tratamento farmacológico , Pneumonia por Pneumocystis/metabolismo , Adulto , Idoso , Antibioticoprofilaxia/métodos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ciclofosfamida/efeitos adversos , Feminino , HIV , Humanos , Hospedeiro Imunocomprometido , Incidência , Linfoma não Hodgkin , Masculino , Pessoa de Meia-Idade , Pneumocystis carinii/patogenicidade , Pneumonia por Pneumocystis/diagnóstico , Prevalência , Estudos Retrospectivos , Rituximab/efeitos adversos , Rituximab/uso terapêutico , Sulfametoxazol/uso terapêutico , Taiwan , Trimetoprima/uso terapêutico , Vincristina/efeitos adversos
10.
Infect Immun ; 85(3)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27993972

RESUMO

We explored differential polarization of macrophages during infection using a rat model of Pneumocystis pneumonia. We observed enhanced pulmonary M1 macrophage polarization in immunosuppressed (IS) hosts, but an M2 predominant response in immunocompetent (IC) hosts following Pneumocystis carinii challenge. Increased inflammation and inducible nitric oxide synthase (iNOS) levels characterized the M1 response. However, macrophage ability to produce nitric oxide was defective. In contrast, the lungs of IC animals revealed a prominent M2 gene signature, and these macrophages effectively elicited an oxidative burst associated with clearance of Pneumocystis In addition, during P. carinii infection the expression of Dectin-1, a critical receptor for recognition and clearance of P. carinii, was upregulated in macrophages of IC animals but suppressed in IS animals. In the absence of an appropriate cytokine milieu for M2 differentiation, Pneumocystis induced an M1 response both in vitro and in vivo The M1 response induced by P. carinii was plastic in nature and reversible with appropriate cytokine stimuli. Finally, we tested whether macrophage polarization can be modulated in vivo and used to help manage the pathogenesis of Pneumocystis pneumonia by adoptive transfer. Treatment with both M1 and M2 cells significantly improved survival of P. carinii-infected IS hosts. However, M2 treatment provided the best outcomes with efficient clearance of P. carinii and reduced inflammation.


Assuntos
Hospedeiro Imunocomprometido , Ativação de Macrófagos/imunologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Pneumocystis/imunologia , Transferência Adotiva , Animais , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos Alveolares/metabolismo , Mortalidade , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/metabolismo , Pneumonia por Pneumocystis/microbiologia , Pneumonia por Pneumocystis/terapia , Ratos
11.
Exp Lung Res ; 42(8-10): 425-439, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27925857

RESUMO

BACKGROUND: Pneumocystis pneumonia is a major cause of morbidity and mortality in patients infected with HIV/AIDS. In this study, we evaluated the intestinal microbial communities associated with the development of experimental Pneumocystis pneumonia, as there is growing evidence that the intestinal microbiota is critical for host defense against fungal pathogens. METHODS: C57BL/6 mice were infected with live Pneumocystis murina (P. murina) via intratracheal inoculation and sacrificed 7 and 14 days postinfection for microbiota analysis. In addition, we evaluated the intestinal microbiota from CD4+ T cell depleted mice infected with P. murina. RESULTS: We found that the diversity of the intestinal microbial community was significantly altered by respiratory infection with P. murina. Specifically, mice infected with P. murina had altered microbial populations, as judged by changes in diversity metrics and relative taxa abundances. We also found that CD4+ T cell depleted mice infected with P. murina exhibited significantly altered intestinal microbiota that was distinct from immunocompetent mice infected with P. murina, suggesting that loss of CD4+ T cells may also affects the intestinal microbiota in the setting of Pneumocystis pneumonia. Finally, we employed a predictive metagenomics approach to evaluate various microbial features. We found that Pneumocystis pneumonia significantly alters the intestinal microbiota's inferred functional potential for carbohydrate, energy, and xenobiotic metabolism, as well as signal transduction pathways. CONCLUSIONS: Our study provides insight into specific-microbial clades and inferred microbial functional pathways associated with Pneumocystis pneumonia. Our data also suggest a role for the gut-lung axis in host defense in the lung.


Assuntos
Microbioma Gastrointestinal , Pneumonia por Pneumocystis/microbiologia , Animais , Metabolismo dos Carboidratos , Metabolismo Energético , Interações Hospedeiro-Patógeno , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia por Pneumocystis/metabolismo , Transdução de Sinais , Xenobióticos
12.
Antimicrob Agents Chemother ; 60(3): 1289-97, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26666941

RESUMO

The combination of all-trans retinoic acid (ATRA) and primaquine (PMQ) has been shown to be effective for therapy of Pneumocystis pneumonia (PCP). Since a high concentration of ATRA has significant adverse effects, the possibility that vitamin D can be used to replace ATRA for PCP therapy was investigated. C57BL/6 mice were immunosuppressed by depleting CD4(+) cells and infected with Pneumocystis murina 1 week after initiation of immunosuppression. Three weeks after infection, the mice were treated orally for 3 weeks with vitamin D3 (VitD3) alone, PMQ alone, a combination of VitD3 and PMQ (VitD3-PMQ), or a combination of trimethoprim and sulfamethoxazole (TMP-SMX). Results showed that VitD3 (300 IU/kg/day) had a synergistic effect with PMQ (5 mg/kg/day) for therapy of PCP. Flow cytometric studies showed that this VitD3-PMQ combination recovered the CD11b(low) CD11c(high) alveolar macrophage population in mice with PCP as effectively as TMP-SMX. The VitD3-PMQ combination also reduced the massive infiltration of inflammatory cells into the lungs and the severity of lung damage. VitD3 was also shown to reduce the dose of TMP-SMX required for effective treatment of PCP. Taken together, results of this study suggest that a VitD3-PMQ combination can be used as an alternative therapy for PCP.


Assuntos
Colecalciferol/farmacologia , Pneumonia por Pneumocystis/tratamento farmacológico , Primaquina/farmacologia , Albuminas/metabolismo , Animais , Antifúngicos/farmacologia , Antígeno CD11b/metabolismo , Cálcio/sangue , Colecalciferol/sangue , Suplementos Nutricionais , Quimioterapia Combinada , Feminino , L-Lactato Desidrogenase/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/patologia , Macrófagos Alveolares/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Pneumocystis/patogenicidade , Pneumonia por Pneumocystis/metabolismo , Pneumonia por Pneumocystis/patologia , Resultado do Tratamento , Combinação Trimetoprima e Sulfametoxazol/administração & dosagem , Combinação Trimetoprima e Sulfametoxazol/farmacologia
13.
J Immunol ; 191(7): 3884-95, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23975863

RESUMO

Although acquired bone marrow failure (BMF) is considered a T cell-mediated autoimmune disease, possible innate immune defects as a cause for systemic immune deviations in response to otherwise innocuous infections have not been extensively explored. In this regard, we recently demonstrated an important role of type I IFNs in protecting hematopoiesis during systemic stress responses to the opportunistic fungal pathogen Pneumocystis in lymphocyte-deficient mice. Mice deficient in both lymphocytes and type I IFN receptor (IFrag(-/-) mice) develop rapidly progressing BMF due to accelerated bone marrow (BM) cell apoptosis associated with innate immune deviations in the BM in response to Pneumocystis lung infection. However, the communication pathway between lung and BM eliciting the induction of BMF in response to this strictly pulmonary infection has been unclear. In this study, we report that absence of an intact type I IFN system during Pneumocystis lung infection not only causes BMF in lymphocyte-deficient mice but also transient BM stress in lymphocyte-competent mice. This is associated with an exuberant systemic IFN-γ response. IFN-γ neutralization prevented Pneumocystis lung infection-induced BM depression in type I IFN receptor-deficient mice and prolonged neutrophil survival time in BM from IFrag(-/-) mice. IL-1ß and upstream regulators of IFN-γ, IL-12, and IL-18 were also upregulated in lung and serum of IFrag(-/-) mice. In conjunction, there was exuberant inflammasome-mediated caspase-1 activation in pulmonary innate immune cells required for processing of IL-18 and IL-1ß. Thus, absence of type I IFN signaling during Pneumocystis lung infection may result in deregulation of inflammasome-mediated pulmonary immune activation, causing systemic immune deviations triggering BMF in this model.


Assuntos
Medula Óssea/metabolismo , Homeostase , Inflamassomos/metabolismo , Interferon Tipo I/metabolismo , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Caspase 1/metabolismo , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Homeostase/efeitos dos fármacos , Homeostase/genética , Interferon gama/antagonistas & inibidores , Interferon gama/sangue , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Pneumocystis/patogenicidade , Receptor de Interferon alfa e beta/genética
14.
Lung ; 191(3): 295-303, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23435873

RESUMO

BACKGROUND: Surfactant protein D (SP-D) is a collection that plays important roles in modulating host defense functions and maintaining phospholipid homeostasis in the lung. The aim of current study was to characterize comparatively the SP-D response in bronchoalveolar lavage (BAL) and serum in three murine models of lung injury, using a validated ELISA technology for estimation of SP-D levels. METHODS: Mice were exposed to lipopolysaccharide, bleomycin, or Pneumocystis carinii (Pc) and sacrificed at different time points. RESULTS: In lipopolysaccharide-challenged mice, the level of SP-D in BAL increased within 6 h, peaked at 51 h (4,518 ng/ml), and returned to base level at 99 h (612 ng/ml). Serum levels of SP-D increased immediately (8.6 ng/ml), peaked at 51 h (16 ng/ml), and returned to base levels at 99 h (3.8 ng/ml). In a subacute bleomycin inflammation model, SP-D levels were 4,625 and 367 ng/ml in BAL and serum, respectively, 8 days after exposure. In a chronic Pc inflammation model, the highest level of SP-D was observed 6 weeks after inoculation, with BAL and serum levels of 1,868 and 335 ng/ml, respectively. CONCLUSIONS: We conclude that serum levels of SP-D increase during lung injury, with a sustained increment during chronic inflammation compared with acute inflammation. A quick upregulation of SP-D in serum in response to acute airway inflammation supports the notion that SP-D translocates from the airways into the vascular system, in favor of being synthesized systemically. The study also confirms the concept of using increased SP-D serum levels as a biomarker of especially chronic airway inflammation.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar/metabolismo , Pulmão/metabolismo , Pneumonia por Pneumocystis/metabolismo , Proteína D Associada a Surfactante Pulmonar/metabolismo , Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/microbiologia , Animais , Bleomicina , Líquido da Lavagem Broncoalveolar/química , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Imuno-Histoquímica , Lipopolissacarídeos , Pulmão/microbiologia , Lesão Pulmonar/sangue , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Pneumocystis carinii , Pneumonia por Pneumocystis/sangue , Pneumonia por Pneumocystis/microbiologia , Proteína D Associada a Surfactante Pulmonar/sangue , Fatores de Tempo
15.
Infect Immun ; 80(11): 3912-20, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22927048

RESUMO

Pneumocystis is an opportunistic fungal pathogen that causes pneumonia in a variety of clinical settings. An early step in Pneumocystis infection involves the attachment of organisms to alveolar epithelial cells (AECs). AECs produce chemokines in response to Pneumocystis stimulation, but the upstream host-pathogen interactions that activate AEC signaling cascades are not well-defined. MyD88 is an adaptor molecule required for activation of proinflammatory signaling cascades following Toll-like receptor (TLR)-dependent recognition of conserved molecular patterns on pathogens. To determine whether the TLR/MyD88 pathway is required for the AEC chemokine response to Pneumocystis, wild-type (WT) and MyD88-deficient AECs were incubated with Pneumocystis. As expected, WT AECs produced CCL2 and CXCL2 following Pneumocystis stimulation. In contrast, MyD88-deficient AECs were severely impaired in their ability to respond to Pneumocystis. MyD88-deficient AECs did not display Pneumocystis-induced Jun N-terminal protein kinase activation and produced much less chemokine than Pneumocystis-stimulated WT AECs. Using a panel of TLR agonists, primary murine AECs were found to respond vigorously to TLR2 and TLR4 agonists. However, the AEC chemokine response to Pneumocystis did not require TLR2 or TLR4. Surprisingly, the interleukin-1 receptor (IL-1R) was required for an AEC chemokine response to Pneumocystis. The role of MyD88 in early responses during Pneumocystis infection was supported by in vivo studies demonstrating that MyD88-deficient mice showed impaired Pneumocystis-stimulated chemokine production and impaired inflammatory cell recruitment. These data indicate an important role for MyD88 in the AEC inflammatory response to Pneumocystis.


Assuntos
Fator 88 de Diferenciação Mieloide/metabolismo , Pneumocystis/imunologia , Pneumonia por Pneumocystis/metabolismo , Alvéolos Pulmonares/citologia , Receptores de Interleucina-1/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Pneumocystis/isolamento & purificação , Pneumonia por Pneumocystis/imunologia , Transdução de Sinais
16.
Am J Pathol ; 181(1): 151-62, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22626807

RESUMO

HIV infection causes loss of CD4(+) T cells and type 1 interferon (IFN)-producing and IFN-responsive dendritic cells, resulting in immunodeficiencies and susceptibility to opportunistic infections, such as Pneumocystis. Osteoporosis and bone marrow failure are additional unexplained complications in HIV-positive patients and patients with AIDS, respectively. We recently demonstrated that mice that lack lymphocytes and IFN a/b receptor (IFrag(-/-)) develop bone marrow failure after Pneumocystis lung infection, whereas lymphocyte-deficient, IFN α/ß receptor-competent mice (RAG(-/-)) had normal hematopoiesis. Interestingly, infected IFrag(-/-) mice also exhibited bone fragility, suggesting loss of bone mass. We quantified bone changes and evaluated the potential connection between progressing bone fragility and bone marrow failure after Pneumocystis lung infection in IFrag(-/-) mice. We found that Pneumocystis infection accelerated osteoclastogenesis as bone marrow failure progressed. This finding was consistent with induction of osteoclastogenic factors, including receptor-activated nuclear factor-κB ligand and the proapoptotic factor tumor necrosis factor-related apoptosis-inducing ligand, in conjunction with their shared decoy receptor osteoprotegerin, in the bone marrow of infected IFrag(-/-) mice. Deregulation of this axis has also been observed in HIV-positive individuals. Biphosphonate treatment of IFrag(-/-) mice prevented bone loss and protected loss of hematopoietic precursor cells that maintained activity in vitro but did not prevent loss of mature neutrophils. Together, these data show that bone loss and bone marrow failure are partially linked, which suggests that the deregulation of the receptor-activated nuclear factor-κB ligand/osteoprotegerin/tumor necrosis factor-related apoptosis-inducing ligand axis may connect the two phenotypes in our model.


Assuntos
Interferon Tipo I/fisiologia , Osteoclastos/fisiologia , Osteoporose/microbiologia , Pneumonia por Pneumocystis/complicações , Síndrome de Resposta Inflamatória Sistêmica/microbiologia , Anemia Aplástica , Animais , Conservadores da Densidade Óssea/uso terapêutico , Medula Óssea/metabolismo , Doenças da Medula Óssea , Transtornos da Insuficiência da Medula Óssea , Diferenciação Celular/fisiologia , Citocinas/metabolismo , Difosfonatos/uso terapêutico , Progressão da Doença , Fêmur/diagnóstico por imagem , Fêmur/patologia , Hemoglobinúria Paroxística/microbiologia , Hemoglobinúria Paroxística/fisiopatologia , Hemoglobinúria Paroxística/prevenção & controle , Camundongos , Camundongos Knockout , Camundongos SCID , Osteoclastos/patologia , Osteoporose/diagnóstico por imagem , Osteoporose/fisiopatologia , Osteoporose/prevenção & controle , Osteoprotegerina/metabolismo , Pneumonia por Pneumocystis/metabolismo , Pneumonia por Pneumocystis/fisiopatologia , Ligante RANK/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/complicações , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Microtomografia por Raio-X
17.
Chin Med J (Engl) ; 124(17): 2683-6, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22040424

RESUMO

BACKGROUND: Pneumocystis jirovecii pneumonia (PCP) is one of the most common and fatal infections in non-AIDS immunocompromised patients, which is difficult to diagnose by traditional morphologic methods. This study evaluated polymerase chain reaction (PCR) assays of Pneumocystis jirovecii mitochondrial large subunits ribosomal RNA in sputum and bronchioalveolar lavage fluid (BALF) for diagnosing PCP. METHODS: Sputum and BALF specimens from two groups were collected: one group (PCP group) included 20 patients definitely diagnosed of PCP by Gomori methenamine silver (GMS) stains of BALF; the other group (non-PCP group) included 40 patients. Each specimen was examined by GMS stains and PCR assays. RESULTS: GMS stains of BALF in PCP group were 100% positive (20/20), GMS stains of sputum in PCP group were 35% positive (7/20); GMS stains of BALF in non-PCP group were 100% negative (40/40), GMS stains of sputum in non-PCP group were 100% negative (40/40). PCR assays of BALF in PCP group were 100% positive (20/20), PCR assays of sputum in PCP group were 100% positive (20/20); PCR assays of BALF in non-PCP group were 100% negative (40/40), PCR assays of sputum in non-PCP group were 100% negative (40/40). Sensitivity and specificity of PCR assays of sputum and BALF were both 100%; positive and negative predictive values were also both 100%. CONCLUSION: The diagnostic value of PCR assays of Pneumocystis jirovecii mitochondrial large subunits ribosomal RNA on sputum and BALF for pneumocystis pneumonia are both high and equivalent.


Assuntos
Síndrome da Imunodeficiência Adquirida/diagnóstico , Pneumonia por Pneumocystis/diagnóstico , Reação em Cadeia da Polimerase/métodos , Síndrome da Imunodeficiência Adquirida/genética , Líquido da Lavagem Broncoalveolar/química , Humanos , Pneumonia por Pneumocystis/genética , Pneumonia por Pneumocystis/metabolismo , Escarro/química
18.
J Immunol ; 186(10): 5956-67, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21471447

RESUMO

We recently demonstrated that lack of type I IFN signaling (IFNAR knockout) in lymphocyte-deficient mice (IFrag(-/-)) results in bone marrow (BM) failure after Pneumocystis lung infection, whereas lymphocyte-deficient mice with intact IFNAR (RAG(-/-)) had normal hematopoiesis. In the current work, we performed studies to define further the mechanisms involved in the induction of BM failure in this system. BM chimera experiments revealed that IFNAR expression was required on BM-derived but not stroma-derived cells to prevent BM failure. Signals elicited after day 7 postinfection appeared critical in determining BM cell fate. We observed caspase-8- and caspase-9-mediated apoptotic cell death, beginning with neutrophils. Death of myeloid precursors was associated with secondary oxidative stress, and decreasing colony-forming activity in BM cell cultures. Treatment with N-acetylcysteine could slow the progression of, but not prevent, BM failure. Type I IFN signaling has previously been shown to expand the neutrophil life span and regulate the expression of some antiapoptotic factors. Quantitative RT-PCR demonstrated reduced mRNA abundance for the antiapoptotic factors BCL-2, IAP2, MCL-1, and others in BM cells from IFrag(-/-) compared with that in BM cells from RAG(-/-) mice at day 7. mRNA and protein for the proapoptotic cytokine TNF-α was increased, whereas mRNA for the growth factors G-CSF and GM-CSF was reduced. In vivo anti-TNF-α treatment improved precursor cell survival and activity in culture. Thus, we propose that lack of type I IFN signaling results in decreased resistance to inflammation-induced proapoptotic stressors and impaired replenishment by precursors after systemic responses to Pneumocystis lung infection. Our finding may have implications in understanding mechanisms underlying regenerative BM depression/failure during complex immune deficiencies such as AIDS.


Assuntos
Apoptose , Células da Medula Óssea/fisiologia , Hematopoese , Interferon Tipo I/imunologia , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/fisiopatologia , Acetilcisteína/farmacologia , Animais , Proteína 3 com Repetições IAP de Baculovírus , Células da Medula Óssea/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Genes RAG-1 , Genes bcl-2 , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Proteínas Inibidoras de Apoptose/genética , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Dados de Sequência Molecular , Proteína de Sequência 1 de Leucemia de Células Mieloides , Células Progenitoras Mieloides/imunologia , Neutrófilos/imunologia , Estresse Oxidativo , Pneumocystis/patogenicidade , Pneumonia por Pneumocystis/metabolismo , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/análise , Quimera por Radiação , Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Ubiquitina-Proteína Ligases
19.
Am J Respir Cell Mol Biol ; 44(4): 540-7, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20558778

RESUMO

The effect of nitric oxide (NO) on Pneumocystis (Pc) organisms, the role of NO in the defense against infection with Pc, and the production of NO by alveolar macrophages (AMs) during Pneumocystis pneumonia (PCP) were investigated. The results indicate that NO was toxic to Pc organisms and inhibited their proliferation in culture. When the production of NO was inhibited by intraperitoneal injection of rats with the nitric oxide synthase inhibitor L-N(5)-(1-iminoethyl) ornithine, progression of Pc infection in immunocompetent rats was enhanced. Concentrations of NO in bronchoalveolar lavage fluids from immunosuppressed, Pc-infected rats and mice were greatly reduced, compared with those from uninfected animals, and AMs from these animals were defective in NO production. However, inducible nitric oxide synthase (iNOS) mRNA and protein concentrations were high in AMs from Pc-infected rats and mice. Immunoblot analysis showed that iNOS in AMs from Pc-infected rats existed primarily as a monomer, but the homo-dimerization of iNOS monomers was required for the production of NO. When iNOS dimerization cofactors, including calmodulin, were added to macrophage lysates, iNOS dimerization increased, whereas incubation of the same lysates with all cofactors except calmodulin did not rescue iNOS dimer formation. These data suggest that NO is important in the defense against Pc infection, but that the production of NO in AMs during PCP is defective because of the reduced dimerization of iNOS.


Assuntos
Macrófagos Alveolares/metabolismo , Óxido Nítrico/biossíntese , Pneumonia por Pneumocystis/metabolismo , Pneumonia por Pneumocystis/patologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Extratos Celulares , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Coenzimas/farmacologia , Meios de Cultura/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/enzimologia , Macrófagos Alveolares/microbiologia , Camundongos , Viabilidade Microbiana/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/metabolismo , Ornitina/farmacologia , Pneumocystis/citologia , Pneumocystis/efeitos dos fármacos , Pneumonia por Pneumocystis/enzimologia , Multimerização Proteica/efeitos dos fármacos , Ratos , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos
20.
Artigo em Chinês | MEDLINE | ID: mdl-24830202

RESUMO

Sprague-Dawley (SD) rat model of Pneumocystis carinii pneumonia (PCP) was established by groin subcutaneous injection with dexamethasone sodium phosphate 35 mg each twice a week for eight weeks. There were two groups: infected group (eighteen rats) and normal control group (six rats) . Pathological changes in lung tissues were observed in the lung imprint after staining with Gomori methenamine silver (GMS) and in tissue sections after staining with hematoxylin-eosin (HE). The activity of superoxide dismutase (SOD) and the content of lipid peroxide (LPO) in lung tissue homogenate were detected by spectrophotometric method. Results showed that in infected group more PC cysts were found in the lung imprint and typical pathological change observed in the lung section. SOD activity in infected group [(31.49 +/- 7.18) U/mgprot] decreased significantly compared with the control [(54.41 +/- 8.97) U/mgprot] (P < 0.01), but LPO in infected group [(2.26 +/- 0.21) nmol/mgprot] was higher significantly than the control [(1.63 +/- 0.01) nmol/mgprot] (P < 0.01).


Assuntos
Peróxidos Lipídicos/metabolismo , Pulmão/patologia , Pneumonia por Pneumocystis/metabolismo , Superóxido Dismutase/metabolismo , Animais , Feminino , Pulmão/metabolismo , Masculino , Pneumonia por Pneumocystis/patologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA