Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros












Intervalo de año de publicación
1.
mBio ; 11(2)2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32127451

RESUMEN

Pneumocystis, a major opportunistic pathogen in patients with a broad range of immunodeficiencies, contains abundant surface proteins encoded by a multicopy gene family, termed the major surface glycoprotein (Msg) gene superfamily. This superfamily has been identified in all Pneumocystis species characterized to date, highlighting its important role in Pneumocystis biology. In this report, through a comprehensive and in-depth characterization of 459 msg genes from 7 Pneumocystis species, we demonstrate, for the first time, the phylogeny and evolution of conserved domains in Msg proteins and provide a detailed description of the classification, unique characteristics, and phylogenetic relatedness of five Msg families. We further describe, for the first time, the relative expression levels of individual msg families in two rodent Pneumocystis species, the substantial variability of the msg repertoires in P. carinii from laboratory and wild rats, and the distinct features of the expression site for the classic msg genes in Pneumocystis from 8 mammalian host species. Our analysis suggests multiple functions for this superfamily rather than just conferring antigenic variation to allow immune evasion as previously believed. This study provides a rich source of information that lays the foundation for the continued experimental exploration of the functions of the Msg superfamily in Pneumocystis biology.IMPORTANCEPneumocystis continues to be a major cause of disease in humans with immunodeficiency, especially those with HIV/AIDS and organ transplants, and is being seen with increasing frequency worldwide in patients treated with immunodepleting monoclonal antibodies. Annual health care associated with Pneumocystis pneumonia costs ∼$475 million dollars in the United States alone. In addition to causing overt disease in immunodeficient individuals, Pneumocystis can cause subclinical infection or colonization in healthy individuals, which may play an important role in species preservation and disease transmission. Our work sheds new light on the diversity and complexity of the msg superfamily and strongly suggests that the versatility of this superfamily reflects multiple functions, including antigenic variation to allow immune evasion and optimal adaptation to host environmental conditions to promote efficient infection and transmission. These findings are essential to consider in developing new diagnostic and therapeutic strategies.


Asunto(s)
Evolución Molecular , Proteínas Fúngicas/genética , Variación Genética , Genoma Fúngico , Glicoproteínas de Membrana/genética , Filogenia , Pneumocystis/genética , Animales , Mamíferos/microbiología , Pneumocystis/clasificación , Ratas , Homología de Secuencia de Ácido Nucleico
2.
Sci Rep ; 7(1): 12307, 2017 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-28951592

RESUMEN

Ethanol-induced hepatic lipophagy plays an important cytoprotective role against liver injury, but its mechanism is not fully determined. In the present study, ethanol-induced lipophagy was studied in an immortalized mouse hepatocyte line, AML12. We found that ethanol treatment elevated lipid content in these cells, which could be regulated by autophagy. To determine the potential mechanism, we investigated the role of a key adaptor molecule SQSTM1/p62. SQSTM1 can bind to LC3 on autophagosomes and ubiquitinated molecules on cargos, thus facilitating the autophagic engulfment of the cargo. We found that both LC3 and SQSTM1 could colocalize with lipid droplets (LDs) following ethanol treatment. Colocalization of LC3 with LDs was significantly inhibited by SQSTM1 knockdown, which also reduced ethanol-induced lipid elevation. In addition, increased ubiquitin signals were found to colocalize with SQSTM1 on LDs in response to ethanol. Moreover, the SQSTM1 signal was colocalized with that of perilipin1, a major protein on LDs. Finally, perilipin1 knockdown significantly altered ethanol-induced lipophagy. Taken together, these data support a model in which autophagosomes were directed to the LDs via SQSTM1, which bound to ubiquitinated proteins, possibly including perilipin 1, on LDs. This study provides a potential mechanistic explanation to how ethanol induces lipophagy in hepatocytes.


Asunto(s)
Autofagia/efectos de los fármacos , Etanol/toxicidad , Hepatocitos/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Proteína Sequestosoma-1/metabolismo , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Hepatocitos/química , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Lípidos/análisis , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Perilipina-1/metabolismo , Proteína Sequestosoma-1/genética , Ubiquitina/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-28760906

RESUMEN

The combination of trimethoprim and sulfamethoxazole (TMP-SMX) is the most effective regimen for therapy of Pneumocystis pneumonia (PCP). As many patients with PCP are allergic or do not respond to it, efforts have been devoted to develop alternative therapies for PCP. We have found that the combination of vitamin D3 (VitD3) (300 IU/kg/day) and primaquine (PMQ) (5 mg/kg/day) was as effective as TMP-SMX for therapy of PCP. In this study, we investigated the mechanisms by which vitamin D enhances the efficacy of PMQ. C57BL/6 mice were immunosuppressed by CD4+ cell depletion, infected with Pneumocystismurina for 8 weeks, and then treated for 9 days with the combination of VitD3 and PMQ (VitD3-PMQ) or with TMP-SMX or PMQ to serve as controls. The results showed that vitamin D supplementation increased the number of CD11c+ cells, suppressed the production of proinflammatory cytokines (tumor necrosis factor alpha [TNF-α], gamma interferon [IFN-γ], and interleukin-6 [IL-6]) and inducible nitric oxide synthase (iNOS), and enhanced the expression of genes related to antioxidation (glutathione reductase and glutamate-cysteine ligase modifier subunit), antimicrobial peptides (cathelicidin), and autophagy (ATG5 and beclin-1). These results suggest that the main action of vitamin D is enhancing the ability of the host to defend against Pneumocystis infection.


Asunto(s)
Antibacterianos/uso terapéutico , Pneumocystis/efectos de los fármacos , Neumonía por Pneumocystis/tratamiento farmacológico , Primaquina/uso terapéutico , Vitamina D/uso terapéutico , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Proteína 5 Relacionada con la Autofagia/biosíntesis , Beclina-1/biosíntesis , Sinergismo Farmacológico , Femenino , Glutamato-Cisteína Ligasa/biosíntesis , Glutatión Reductasa/biosíntesis , Humanos , Interferón gamma/biosíntesis , Interleucina-6/biosíntesis , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Neumonía por Pneumocystis/microbiología , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico , Factor de Necrosis Tumoral alfa/biosíntesis , Catelicidinas
4.
Am J Pathol ; 186(9): 2310-6, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27392970

RESUMEN

Idiopathic pulmonary fibrosis is a devastating disease, with no good diagnostic biomarker and limited treatment options. Previous studies suggest that collagen V overexpression and collagen V-mediated immune response play roles in the pathogenesis of idiopathic pulmonary fibrosis. This study aimed to identify dysregulated miRNA-related collagen V overexpression during idiopathic pulmonary fibrosis. We found that the expression levels of miR-185 and miR-186 were decreased in the lungs of idiopathic pulmonary fibrosis patients. The levels of miR-185 and miR-186 were not correlated with disease severity of idiopathic pulmonary fibrosis. The direct regulation of COL5A1 by miR-185 and miR-186 was confirmed by a luciferase reporter assay. Furthermore, mimics of miR-185 and miR-186 blocked transforming growth factor-ß-induced collagen V overexpression and alleviated transforming growth factor-ß-induced epithelial-mesenchymal transition in A549 cells and HCC827 cells. Our findings suggest that attenuated expression of miR-185 and miR-186 may be responsible for collagen V overexpression during idiopathic pulmonary fibrosis, and these miRNAs may serve as pathogenesis-related biomarkers and treatment targets.


Asunto(s)
Colágeno Tipo V/biosíntesis , Transición Epitelial-Mesenquimal/genética , Fibrosis Pulmonar Idiopática/patología , MicroARNs/metabolismo , Anciano , Western Blotting , Femenino , Regulación de la Expresión Génica/genética , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Antimicrob Agents Chemother ; 60(3): 1289-97, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26666941

RESUMEN

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.


Asunto(s)
Colecalciferol/farmacología , Neumonía por Pneumocystis/tratamiento farmacológico , Primaquina/farmacología , Albúminas/metabolismo , Animales , Antifúngicos/farmacología , Antígeno CD11b/metabolismo , Calcio/sangre , Colecalciferol/sangre , Suplementos Dietéticos , Quimioterapia Combinada , Femenino , L-Lactato Deshidrogenasa/metabolismo , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/efectos de los fármacos , Ratones Endogámicos C57BL , Pneumocystis/patogenicidad , Neumonía por Pneumocystis/metabolismo , Neumonía por Pneumocystis/patología , Resultado del Tratamiento , Combinación Trimetoprim y Sulfametoxazol/administración & dosificación , Combinación Trimetoprim y Sulfametoxazol/farmacología
6.
Med Mycol ; 53(8): 868-73, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26337088

RESUMEN

The performance characteristics of the recently available analyte-specific reagent based enzyme immunoassay (ASR-EIA) and in vitro diagnostic (IVD) kit for urine Histoplasma antigen detection were evaluated in a cohort of 50 clinically characterized patients with histoplasmosis and 50 control patients. Overall sensitivity and specificity of the ASR-EIA were significantly improved compared with those of the IVD kit (sensitivity 72% vs. 22%, P<.001, specificity 98% vs. 84%, P = .014). Fourteen specimens from patients with clinically characterized histoplasmosis (five with pulmonary histoplasmosis and nine with progressive disseminated histoplasmosis) were falsely negative by ASR-EIA. All 10 specimens from patients with severe symptoms of progressive disseminated histoplasmosis were positive by ASR-EIA, although the average reading value of these 10 specimens was not significantly different from that of others with positive results. Compared to the MiraVista antigen assay, both the IVD kit and the ASR-EIA were significantly less sensitive in detecting Histoplasma antigen in the urine of patients with histoplasmosis. The ASR-EIA and MiraVista assay had comparable specificity. In conclusion, the ASR-EIA has improved performance compared with the IVD kit in the detection of Histoplasma antigen in the urine. However, users should be aware of the potential for false negative results using the currently recommended cutoff value.


Asunto(s)
Histoplasmosis/diagnóstico , Juego de Reactivos para Diagnóstico , Pruebas Serológicas/métodos , Antígenos Fúngicos/orina , Estudios de Cohortes , Histoplasma/inmunología , Humanos , Técnicas para Inmunoenzimas/métodos , Sensibilidad y Especificidad
7.
Infect Immun ; 83(2): 572-82, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25404033

RESUMEN

Myeloid-derived suppressor cells (MDSCs) were recently found to accumulate in the lungs during Pneumocystis pneumonia (PcP). Adoptive transfer of these cells caused lung damage in recipient mice, suggesting that MDSC accumulation is a mechanism of pathogenesis in PcP. In this study, the phagocytic activity of alveolar macrophages (AMs) was found to decrease by 40% when they were incubated with MDSCs from Pneumocystis-infected mice compared to those incubated with Gr-1(+) cells from the bone marrow of uninfected mice. The expression of the PU.1 gene in AMs incubated with MDSCs also was decreased. This PU.1 downregulation was due mainly to decreased histone 3 acetylation and increased DNA methylation caused by MDSCs. MDSCs were found to express high levels of PD-L1, and alveolar macrophages (AMs) were found to express high levels of PD-1 during PcP. Furthermore, PD-1 expression in AMs from uninfected mice was increased by 18-fold when they were incubated with MDSCs compared to those incubated with Gr-1(+) cells from the bone marrow of uninfected mice. The adverse effects of MDSCs on AMs were diminished when the MDSCs were pretreated with anti-PD-L1 antibody, suggesting that MDSCs disable AMs through PD-1/PD-L1 ligation during PcP.


Asunto(s)
Antígeno B7-H1/inmunología , Macrófagos Alveolares/inmunología , Células Mieloides/inmunología , Neumonía por Pneumocystis/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Acetilación , Animales , Antígeno B7-H1/biosíntesis , Metilación de ADN , Regulación hacia Abajo , Histonas/metabolismo , Pulmón/citología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Fagocitosis/inmunología , Pneumocystis carinii/inmunología , Pneumocystis carinii/patogenicidad , Neumonía por Pneumocystis/patología , Receptor de Muerte Celular Programada 1/biosíntesis , Receptor de Muerte Celular Programada 1/genética , Proteínas Proto-Oncogénicas/biosíntesis , Receptores de Quimiocina/biosíntesis , Transactivadores/biosíntesis
8.
PLoS One ; 8(1): e53479, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23308231

RESUMEN

Pneumocystis pneumonia (PcP) develops in immunocompromised patients. Alveolar macrophages play a key role in the recognition, phagocytosis, and degradation of Pneumocystis, but their number is decreased in PcP. Our study of various inflammatory components during PcP found that myeloid-derived suppressor cells (MDSCs) accumulate in the lungs of mice and rats with Pneumocystis pneumonia (PcP). We hypothesized that treatment with all-trans retinoic acid (ATRA), a metabolite of vitamin A, may effectively control Pneumocystis (Pc) infection by inducing MDSCs to differentiate to AMs. In rodent models of PcP, we found that 5 weeks of ATRA treatment reduced the number of MDSCs in the lungs and increased the number of AMs which cleared Pc infection. We also found that ATRA in combination with primaquine was as effective as the combination of trimethoprim and sulfamethaxazole for treatment of PcP and completely eliminated MDSCs and Pc organisms in the lungs in two weeks. No relapse of PcP was seen after three weeks of the ATRA-primaquine combination treatment. Prolonged survival of Pc-infected animals was also achieved by this regimen. This is the very first successful development of a therapeutic regimen for PcP that combines an immune modulator with an antibiotic, enabling the hosts to effectively defend the infection. Results of our study may serve as a model for development of novel therapies for other infections with MDSC accumulation.


Asunto(s)
Pneumocystis/efectos de los fármacos , Neumonía por Pneumocystis/tratamiento farmacológico , Primaquina/farmacología , Tretinoina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia Combinada/métodos , Femenino , Humanos , Huésped Inmunocomprometido , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos BALB C , Pneumocystis/fisiología , Neumonía por Pneumocystis/inmunología , Neumonía por Pneumocystis/microbiología , Neumonía por Pneumocystis/mortalidad , Ratas , Ratas Sprague-Dawley , Análisis de Supervivencia , Combinación Trimetoprim y Sulfametoxazol/farmacología
9.
Infect Immun ; 80(10): 3634-41, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22868498

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of hematopoietic precursors with the ability to adversely affect host immunity. They have been shown to accumulate in pathological conditions, such as cancer and some microbial diseases. In the mouse and rat models of Pneumocystis pneumonia (PcP), we found a distinct population of cells with MDSC-like morphology in the bronchoalveolar lavage (BAL) fluid, constituting up to 50% of the total cells in BAL fluid. These cells were not seen in the BAL fluid from normal animals or from Pneumocystis-infected animals that had been successfully treated for PcP with a combination of trimethoprim and sulfamethoxazole. With flow cytometry, these cells were found to express the characteristic MDSC surface markers Gr-1 and CD11b in mice or CD11bc and His48 in rats. Using reverse transcription-PCR, we demonstrated that these cells produced high levels of arginase-1 and inducible nitric oxide synthase (iNOS) mRNA. These cells were shown to suppress CD4(+) T-cell proliferation in response to stimulation by anti-CD3 and anti-CD28 antibodies. Adoptive transfer of these cells to normal mice caused lung damage, as indicated by elevated levels of albumin and lactate dehydrogenase in the BAL fluid. These experiments provide evidence of the presence of MDSCs in the lungs during PcP. Further studies on the roles of MDSCs in PcP are warranted in order to develop treatment strategies which can reduce the number of MDSCs and the damage caused by these cells.


Asunto(s)
Células Madre Hematopoyéticas/citología , Pulmón/citología , Neumonía por Pneumocystis/inmunología , Albúminas/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Antígeno CD11b/genética , Proliferación Celular , Femenino , Citometría de Flujo , Regulación de la Expresión Génica/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunidad Celular , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Linfocitos T/citología , Linfocitos T/fisiología
10.
Am J Respir Cell Mol Biol ; 44(4): 540-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20558778

RESUMEN

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.


Asunto(s)
Macrófagos Alveolares/metabolismo , Óxido Nítrico/biosíntesis , Neumonía por Pneumocystis/metabolismo , Neumonía por Pneumocystis/patología , Animales , Líquido del Lavado Bronquioalveolar/citología , Extractos Celulares , Línea Celular , Proliferación Celular/efectos de los fármacos , Coenzimas/farmacología , Medios de Cultivo/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/microbiología , Ratones , Viabilidad Microbiana/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Ornitina/farmacología , Pneumocystis/citología , Pneumocystis/efectos de los fármacos , Neumonía por Pneumocystis/enzimología , Multimerización de Proteína/efectos de los fármacos , Ratas , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
11.
J Biol Chem ; 285(51): 40322-32, 2010 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-20956540

RESUMEN

A decreased clearance of apoptotic cells (efferocytosis) by alveolar macrophages (AM) may contribute to inflammation in emphysema. The up-regulation of ceramides in response to cigarette smoking (CS) has been linked to AM accumulation and increased detection of apoptotic alveolar epithelial and endothelial cells in lung parenchyma. We hypothesized that ceramides inhibit the AM phagocytosis of apoptotic cells. Release of endogenous ceramides via sphingomyelinase or exogenous ceramide treatments dose-dependently impaired apoptotic Jurkat cell phagocytosis by primary rat or human AM, irrespective of the molecular species of ceramide. Similarly, in vivo augmentation of lung ceramides via intratracheal instillation in rats significantly decreased the engulfment of instilled target apoptotic thymocytes by resident AM. The mechanism of ceramide-induced efferocytosis impairment was dependent on generation of sphingosine via ceramidase. Sphingosine treatment recapitulated the effects of ceramide, dose-dependently inhibiting apoptotic cell clearance. The effect of ceramide on efferocytosis was associated with decreased membrane ruffle formation and attenuated Rac1 plasma membrane recruitment. Constitutively active Rac1 overexpression rescued AM efferocytosis against the effects of ceramide. CS exposure significantly increased AM ceramides and recapitulated the effect of ceramides on Rac1 membrane recruitment in a sphingosine-dependent manner. Importantly, CS profoundly inhibited AM efferocytosis via ceramide-dependent sphingosine production. These results suggest that excessive lung ceramides may amplify lung injury in emphysema by causing both apoptosis of structural cells and inhibition of their clearance by AM.


Asunto(s)
Apoptosis/efectos de los fármacos , Ceramidas/farmacología , Macrófagos Alveolares/metabolismo , Fumar/efectos adversos , Animales , Membrana Celular/metabolismo , Membrana Celular/patología , Ceramidasas/metabolismo , Ceramidas/metabolismo , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Células Jurkat , Macrófagos Alveolares/patología , Masculino , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Proteína de Unión al GTP rac1/metabolismo
13.
BMC Microbiol ; 10: 103, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20377877

RESUMEN

BACKGROUND: Pneumocystis pneumonia is a common opportunistic disease in AIDS patients. The alveolar macrophage is an important effector cell in the clearance of Pneumocystis organisms by phagocytosis. However, both the number and phagocytic activity of alveolar macrophages are decreased in Pneumocystis infected hosts. To understand how Pneumocystis inactivates alveolar macrophages, Affymetrix GeneChip RG-U34A DNA microarrays were used to study the difference in global gene expression in alveolar macrophages from uninfected and Pneumocystis carinii-infected Sprague-Dawley rats. RESULTS: Analyses of genes that were affected by Pneumocystis infection showed that many functions in the cells were affected. Antigen presentation, cell-mediated immune response, humoral immune response, and inflammatory response were most severely affected, followed by cellular movement, immune cell trafficking, immunological disease, cell-to-cell signaling and interaction, cell death, organ injury and abnormality, cell signaling, infectious disease, small molecular biochemistry, antimicrobial response, and free radical scavenging. Since rats must be immunosuppressed in order to develop Pneumocystis infection, alveolar macrophages from four rats of the same sex and age that were treated with dexamethasone for the entire eight weeks of the study period were also examined. With a filter of false-discovery rate less than 0.1 and fold change greater than 1.5, 200 genes were found to be up-regulated, and 144 genes were down-regulated by dexamethasone treatment. During Pneumocystis pneumonia, 115 genes were found to be up- and 137 were down-regulated with the same filtering criteria. The top ten genes up-regulated by Pneumocystis infection were Cxcl10, Spp1, S100A9, Rsad2, S100A8, Nos2, RT1-Bb, Lcn2, RT1-Db1, and Srgn with fold changes ranging between 12.33 and 5.34; and the top ten down-regulated ones were Lgals1, Psat1, Tbc1d23, Gsta1, Car5b, Xrcc5, Pdlim1, Alcam, Cidea, and Pkib with fold changes ranging between -4.24 and -2.25. CONCLUSIONS: In order to survive in the host, Pneumocystis organisms change the expression profile of alveolar macrophages. Results of this study revealed that Pneumocystis infection affects many cellular functions leading to reduced number and activity of alveolar macrophages during Pneumocystis pneumonia.


Asunto(s)
Regulación de la Expresión Génica , Macrófagos Alveolares/fisiología , Infecciones por Pneumocystis/genética , Pneumocystis carinii/fisiología , Animales , Muerte Celular/genética , Dexametasona/farmacología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica/métodos , Inmunidad/genética , Inflamación/genética , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Fagocitosis/genética , Infecciones por Pneumocystis/inmunología , Infecciones por Pneumocystis/metabolismo , Análisis de Componente Principal , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
14.
Infect Immun ; 78(3): 1058-65, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20065023

RESUMEN

Dectin-1 is an important macrophage phagocytic receptor recognizing fungal beta-glucans. In this study, the mRNA levels of the Dectin-1 gene were found to be decreased by 61% in alveolar macrophages (AMs) from Pneumocystis-infected mice. The expression of Dectin-1 protein on the surface of these cells was also significantly decreased. By fluorescence in situ hybridization, mRNA expression levels of the transcription factor PU.1 were also found to be significantly reduced in AMs from Pneumocystis-infected mice. Electrophoretic mobility shift assay showed that PU.1 protein bound Dectin-1 gene promoter. With a luciferase reporter gene driven by the Dectin-1 gene promoter, the expression of the PU.1 gene in NIH 3T3 cells was found to enhance the luciferase activity in a dose-dependent manner. PU.1 expression knockdown by small interfering RNA (siRNA) caused a 63% decrease in Dectin-1 mRNA level and 40% decrease in protein level in AMs. Results of this study indicate that downregulation of PU.1 during Pneumocystis pneumonia leads to decreased expression of Dectin-1 in AMs.


Asunto(s)
Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Proteínas de la Membrana/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Pneumocystis/inmunología , Pneumocystis/patogenicidad , Neumonía por Pneumocystis/inmunología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Células 3T3 , Animales , Fusión Artificial Génica , ADN/metabolismo , Regulación hacia Abajo , Ensayo de Cambio de Movilidad Electroforética , Femenino , Perfilación de la Expresión Génica , Genes Reporteros , Lectinas Tipo C , Luciferasas/biosíntesis , Luciferasas/genética , Macrófagos Alveolares/química , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Ratones , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo
15.
Antimicrob Agents Chemother ; 53(12): 5259-64, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19805570

RESUMEN

Polyamine levels are greatly increased in alveolar macrophages (AMs) during Pneumocystis pneumonia (PCP), leading to increased production of H(2)O(2), which causes AMs to undergo apoptosis. One of the mechanisms by which polyamine levels in AMs are elevated is enhanced uptake of exogenous polyamines. In this study, the possibility of targeting polyamine uptake as a treatment for PCP was examined. Four anthracene- and one benzene-polyamine conjugates that are potential polyamine transport inhibitors, including N1-anthracen-9-ylmethyl-butane-1,4-diamine; N-(4-aminobutyl)-N-anthracen-9-ylmethylbutane-1,4-diamine; N-[4-(4-aminobutylamino)butyl]-N-anthracen-9-ylmethylbutane-1,4-diamine; N-(4-amino-butyl)-N'-(10-[[4-(4-amino-butylamino)butylamino]-methyl]anthracen-9-ylmethyl)butane-1,4-diamine (44-Ant-44); and benzene-polyamine conjugate N-(4-amino-butyl)-N'-(4-[[4-(4-amino-butylamino)butylamino]-methyl]benzyl)butane-1,4-diamine (44-Bn-44), were tested. Compounds 44-Ant-44 and 44-Bn-44 were found to have a very low toxicity to AMs in vitro and were evaluated for their therapeutic effect on PCP in vivo. Sprague-Dawley rats infected with P. carinii for 28 days were intranasally instilled with 50 microl of a 1 mM solution of 44-Bn-44 or 44-Ant-44 every 2 days. Twenty-one days after initiation of the treatment, three to five rats from each group were sacrificed and examined for lung pathology, organism burden, and apoptosis of AMs. Both 44-Bn-44 and 44-Ant-44 reduced organism burdens; however, only 44-Ant-44 decreased the severity of the infection with reduced lung inflammation, increased clearance of exudates, increased air space, and decreased apoptosis of AMs. 44-Ant-44 also significantly prolonged the survival of treated animals. These results suggest that polyamine uptake is a potential target for treatment of PCP.


Asunto(s)
Antifúngicos/uso terapéutico , Transporte Biológico/efectos de los fármacos , Neumonía por Pneumocystis/tratamiento farmacológico , Neumonía por Pneumocystis/microbiología , Poliaminas/metabolismo , Animales , Antracenos/química , Antifúngicos/efectos adversos , Antifúngicos/química , Antifúngicos/farmacología , Apoptosis/efectos de los fármacos , Benceno/química , Femenino , Inyecciones Intramusculares , Macrófagos Alveolares/citología , Macrófagos Alveolares/efectos de los fármacos , Estructura Molecular , Pneumocystis carinii/efectos de los fármacos , Neumonía por Pneumocystis/patología , Ratas , Ratas Sprague-Dawley
16.
Infect Immun ; 77(8): 3344-54, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19487471

RESUMEN

Pneumocystis infection causes increased intracellular levels of reactive oxygen species (ROS) and the subsequent apoptosis of alveolar macrophages (Amø). Assessments of key prosurvival molecules in Amø and bronchoalveolar lavage fluids from infected rats and mice showed low levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) and reduced activation of phosphoinositide-3 kinase (PI-3K). Ubiquitous calcium-sensing protein calmodulin protein and mRNA levels were also reduced in Amø during Pneumocystis pneumonia (Pcp). Calmodulin has been implicated in control of GM-CSF production and PI-3K activation in other immune cell types. Experiments to determine the control of GM-CSF and PI-3K by calmodulin in Amø showed that GM-CSF expression and PI-3K activation could not be induced when calmodulin was inhibited. Calmodulin inhibition also led to increased levels of ROS and apoptosis in cells exposed to bronchoalveolar lavage fluids from infected animals. Supplementation of Amø with exogenous calmodulin increased survival signaling via GM-CSF and PI-3K and reduced ROS and apoptosis. These data support the hypotheses that calmodulin levels at least partially control survival signaling in Amø and that restoration of GM-CSF or PI-3K signaling will improve host response to the organism.


Asunto(s)
Calmodulina/inmunología , Macrófagos Alveolares/inmunología , Neumonía por Pneumocystis/inmunología , Animales , Apoptosis , Supervivencia Celular , Células Cultivadas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Macrófagos Alveolares/química , Ratones , Ratones Endogámicos BALB C , Fosfatidilinositol 3-Quinasas/biosíntesis , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/análisis
17.
J Biol Chem ; 284(12): 8174-84, 2009 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-19158080

RESUMEN

Pneumocystis pneumonia (PcP) is the most common opportunistic disease in immunocompromised patients. Alveolar macrophages are responsible for the clearance of Pneumocystis organisms; however, they undergo a high rate of apoptosis during PcP due to increased intracellular polyamine levels. In this study, the sources of polyamines and mechanisms of polyamine increase and polyamine-induced apoptosis were investigated. The level of ornithine decarboxylase (ODC) was elevated in alveolar macrophages, and the number of alveolar macrophages that took up exogenous polyamines was increased 20-fold during PcP. Monocytes, B lymphocytes, and CD8+ T lymphocytes that were recruited into the lung during PcP expressed high levels of ornithine decarboxylase, suggesting that these cells are sources of polyamines. Both protein and mRNA levels of antizyme inhibitor (AZI) were increased in alveolar macrophages during PcP. This AZI overexpression correlated with increased polyamine uptake by alveolar macrophages, because AZI expression knockdown decreased the polyamine uptake ability of these cells. AZI expression knockdown also decreased the apoptosis rate of alveolar macrophages. Pneumocystis organisms and zymosan A were found to induce AZI overexpression in alveolar macrophages, suggesting that beta-glucan, which is the major component of the Pneumocystis cell wall, induces AZI overexpression. The levels of mRNA, protein, and activity of polyamine oxidase were increased in alveolar macrophages during PcP, indicating that the H(2)O(2) generated during polyamine catabolism caused alveolar macrophages to undergo apoptosis. Taken together, results of this study indicate that Pneumocystis organisms induce AZI overexpression in alveolar macrophages, leading to increased polyamine synthesis and uptake and apoptosis rate of these cells.


Asunto(s)
Apoptosis , Proteínas Portadoras/biosíntesis , Regulación de la Expresión Génica , Macrófagos Alveolares/metabolismo , Pneumocystis carinii , Neumonía por Pneumocystis/metabolismo , Poliaminas/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas Portadoras/inmunología , Pared Celular/inmunología , Pared Celular/metabolismo , Humanos , Peróxido de Hidrógeno/inmunología , Peróxido de Hidrógeno/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/fisiología , Masculino , Ornitina Descarboxilasa/inmunología , Ornitina Descarboxilasa/metabolismo , Inhibidores de la Ornitina Descarboxilasa , Neumonía por Pneumocystis/inmunología , Neumonía por Pneumocystis/patología , Poliaminas/inmunología , Ratas , Ratas Sprague-Dawley , beta-Glucanos/inmunología , beta-Glucanos/metabolismo
18.
Hong Kong; Kompass Books; 2009. 470 p.
Monografía en Inglés | LILACS, ColecionaSUS | ID: biblio-939347
19.
J Formos Med Assoc ; 107(11): 830-42, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18971152

RESUMEN

Pneumocystis pneumonia (PcP) in humans is caused by Pneumocystis jirovecii, which has recently been reclassified as a fungus because its cell wall composition and nucleotide sequences are more similar to those of fungi. PcP occurs only in immunocompromised individuals such as those with AIDS. Despite the use of highly active antiretroviral therapy, PcP remains the leading opportunistic infection in AIDS patients. Based on nucleotide sequence variations in the internal transcribed spacer region of rRNA genes, more than 60 different types of P. jirovecii have been identified. Although type differences do not appear to correlate with the clinical characteristics of PcP, nucleotide sequence variations of the organism have been useful in epidemiologic studies. As a result, some recurrent infections are found to be due to re-infection with new types, and outbreaks due to the same types of P. jirovecii have been identified. Initial diagnosis of PcP is usually based on symptoms and chest radiography. A characteristic histopathologic feature is the presence of acellular eosinophilic exudates and organisms in the alveoli. Ultimate diagnosis of PcP is achieved by demonstration of the organism in induced sputum or bronchoalveolar lavage fluid by tinctorial staining or polymerase chain reaction (PCR). Among the many different PCR methods, the nested PCR that targets the large subunit mitochondrial rRNA gene is the most sensitive and specific. Combination of trimethoprim and sulfamethoxazole is the first choice of drugs for both treatment and prophylaxis of PcP. Other drugs that can be used include a combination of primaquine and clindamycin, pentamidine, atovaquone, and a combination of dapsone and trimethoprim. Pneumocystis organisms have the ability to inactivate the phagocytic activity of alveolar macrophages and to induce them to undergo apoptosis. This apoptosis is due to activation of caspase 9 by polyamines that are present in high levels in the lung and alveolar macrophages during PcP.


Asunto(s)
Neumonía por Pneumocystis , Humanos , Neumonía por Pneumocystis/diagnóstico , Neumonía por Pneumocystis/etiología , Neumonía por Pneumocystis/terapia
20.
Microbes Infect ; 10(4): 334-41, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18400546

RESUMEN

Pneumocystis pneumonia (PcP) is marked by substantial inflammatory damage to the lung. We have found that Toll-like receptor 2 (TLR2) mediates macrophage inflammatory responses to Pneumocystis and hypothesized that TLR2 deficiency would lead to less severe inflammation and milder lung injury during PcP. Histopathology examination showed that TLR2-/- mice with PcP indeed exhibited milder pulmonary inflammation. TLR2-/- mouse lungs contained less TNF-alpha and displayed lower levels of NF-kappaB activation during PcP. However, TLR2-/- mice with PcP displayed increased severity in symptoms and organism burden. The increased organism burden is likely due to defects in protective mechanisms in TLR2-/- mice. mRNA levels of the inducible nitric oxide synthase and NADPH oxidase p47phox, as well as nitric oxide levels in the lungs, were decreased in TLR2-/- PcP mice. Taken together, this study shows that TLR2-mediated inflammatory responses contribute to a certain degree to the clearance of Pneumocystis organism in mice.


Asunto(s)
Inflamación/patología , Pulmón/patología , Neumonía por Pneumocystis/inmunología , Neumonía por Pneumocystis/patología , Receptor Toll-Like 2/inmunología , Animales , Femenino , Pulmón/química , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/biosíntesis , FN-kappa B/análisis , Óxido Nítrico/análisis , Óxido Nítrico Sintasa/biosíntesis , Pneumocystis carinii/aislamiento & purificación , Neumonía por Pneumocystis/fisiopatología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Factor de Necrosis Tumoral alfa/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...