Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 19(8): e1011573, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37624851

RESUMO

Pseudomonas aeruginosa (P.a.) infection accounts for nearly 20% of all cases of hospital acquired pneumonia with mortality rates >30%. P.a. infection induces a robust inflammatory response, which ideally enhances bacterial clearance. Unfortunately, excessive inflammation can also have negative effects, and often leads to cardiac dysfunction with associated morbidity and mortality. However, it remains unclear how P.a. lung infection causes cardiac dysfunction. Using a murine pneumonia model, we found that P.a. infection of the lungs led to severe cardiac left ventricular dysfunction and electrical abnormalities. More specifically, we found that neutrophil recruitment and release of S100A8/A9 in the lungs activates the TLR4/RAGE signaling pathways, which in turn enhance systemic inflammation and subsequent cardiac dysfunction. Paradoxically, global deletion of S100A8/A9 did not improve but aggravated cardiac dysfunction and mortality likely due to uncontrolled bacterial burden in the lungs and heart. Our results indicate that P.a. infection induced release of S100A8/9 is double-edged, providing increased risk for cardiac dysfunction yet limiting P.a. growth.


Assuntos
Cardiopatias , Infecções por Pseudomonas , Animais , Camundongos , Pseudomonas aeruginosa , Coração , Inflamação , Pulmão
2.
J Immunol ; 206(6): 1284-1296, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33568400

RESUMO

Neutralizing Abs suppress HIV infection by accelerating viral clearance from blood circulation in addition to neutralization. The elimination mechanism is largely unknown. We determined that human liver sinusoidal endothelial cells (LSEC) express FcγRIIb as the lone Fcγ receptor, and using humanized FcγRIIb mouse, we found that Ab-opsonized HIV pseudoviruses were cleared considerably faster from circulation than HIV by LSEC FcγRIIb. Compared with humanized FcγRIIb-expressing mice, HIV clearance was significantly slower in FcγRIIb knockout mice. Interestingly, a pentamix of neutralizing Abs cleared HIV faster compared with hyperimmune anti-HIV Ig (HIVIG), although the HIV Ab/Ag ratio was higher in immune complexes made of HIVIG and HIV than pentamix and HIV. The effector mechanism of LSEC FcγRIIb was identified to be endocytosis. Once endocytosed, both Ab-opsonized HIV pseudoviruses and HIV localized to lysosomes. This suggests that clearance of HIV, endocytosis, and lysosomal trafficking within LSEC occur sequentially and that the clearance rate may influence downstream events. Most importantly, we have identified LSEC FcγRIIb-mediated endocytosis to be the Fc effector mechanism to eliminate cell-free HIV by Abs, which could inform development of HIV vaccine and Ab therapy.


Assuntos
Anticorpos Neutralizantes/metabolismo , Endocitose/imunologia , Células Endoteliais/imunologia , Infecções por HIV/imunologia , Receptores de IgG/metabolismo , Animais , Capilares/citologia , Capilares/imunologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Células HEK293 , HIV/imunologia , Infecções por HIV/sangue , Infecções por HIV/patologia , Infecções por HIV/virologia , Voluntários Saudáveis , Humanos , Fígado/irrigação sanguínea , Fígado/imunologia , Lisossomos/metabolismo , Lisossomos/virologia , Masculino , Camundongos , Camundongos Knockout , Cultura Primária de Células , Receptores de IgG/genética
3.
Proc Natl Acad Sci U S A ; 116(37): 18607-18612, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31451661

RESUMO

Influenza virus can disseminate from the lungs to the heart in severe infections and can induce cardiac pathology, but this has been difficult to study due to a lack of small animal models. In humans, polymorphisms in the gene encoding the antiviral restriction factor IFN-induced transmembrane protein 3 (IFITM3) are associated with susceptibility to severe influenza, but whether IFITM3 deficiencies contribute to cardiac dysfunction during infection is unclear. We show that IFITM3 deficiency in a new knockout (KO) mouse model increases weight loss and mortality following influenza virus infections. We investigated this enhanced pathogenesis with the A/PR/8/34 (H1N1) (PR8) influenza virus strain, which is lethal in KO mice even at low doses, and observed increased replication of virus in the lungs, spleens, and hearts of KO mice compared with wild-type (WT) mice. Infected IFITM3 KO mice developed aberrant cardiac electrical activity, including decreased heart rate and irregular, arrhythmic RR (interbeat) intervals, whereas WT mice exhibited a mild decrease in heart rate without irregular RR intervals. Cardiac electrical dysfunction in PR8-infected KO mice was accompanied by increased activation of fibrotic pathways and fibrotic lesions in the heart. Infection with a sublethal dose of a less virulent influenza virus strain (A/WSN/33 [H1N1]) resulted in a milder cardiac electrical dysfunction in KO mice that subsided as the mice recovered. Our findings reveal an essential role for IFITM3 in limiting influenza virus replication and pathogenesis in heart tissue and establish IFITM3 KO mice as a powerful model for studying mild and severe influenza virus-induced cardiac dysfunction.


Assuntos
Cardiopatias/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/genética , Proteínas de Membrana/genética , Miocárdio/patologia , Animais , Modelos Animais de Doenças , Ecocardiografia , Eletrocardiografia , Fibrose , Predisposição Genética para Doença , Coração/diagnóstico por imagem , Coração/virologia , Cardiopatias/diagnóstico , Cardiopatias/patologia , Cardiopatias/virologia , Humanos , Vírus da Influenza A Subtipo H1N1/patogenicidade , Influenza Humana/complicações , Influenza Humana/imunologia , Influenza Humana/virologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Índice de Gravidade de Doença , Replicação Viral/genética , Replicação Viral/imunologia
4.
J Immunol ; 203(8): 2252-2264, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31511357

RESUMO

The elderly population is more susceptible to pulmonary infections, including tuberculosis. In this article, we characterize the impact of aging on the phenotype of mouse alveolar macrophages (AMs) and their response to Mycobacterium tuberculosis. Uninfected AMs were isolated from bronchoalveolar lavage of young (3 mo) and old (18 mo) C57BL/6 mice. AMs from old mice expressed higher mRNA levels of CCL2, IFN-ß, IL-10, IL-12p40, TNF-α, and MIF than young mice, and old mice contained higher levels of CCL2, IL-1ß, IFN-ß, and MIF in their alveolar lining fluid. We identified two distinct AM subpopulations, a major CD11c+ CD11b- population and a minor CD11c+ CD11b+ population; the latter was significantly increased in old mice (4-fold). Expression of CD206, TLR2, CD16/CD32, MHC class II, and CD86 was higher in CD11c+ CD11b+ AMs, and these cells expressed monocytic markers Ly6C, CX3CR1, and CD115, suggesting monocytic origin. Sorted CD11c+ CD11b+ AMs from old mice expressed higher mRNA levels of CCL2, IL-1ß, and IL-6, whereas CD11c+ CD11b- AMs expressed higher mRNA levels of immune-regulatory cytokines IFN-ß and IL-10. CD11c+ CD11b+ AMs phagocytosed significantly more M. tuberculosis, which expressed higher RNA levels of genes required for M. tuberculosis survival. Our studies identify two distinct AM populations in old mice: a resident population and an increased CD11c+ CD11b+ AM subpopulation expressing monocytic markers, a unique inflammatory signature, and enhanced M. tuberculosis phagocytosis and survival when compared with resident CD11c+ CD11b- AMs, which are more immune regulatory in nature.


Assuntos
Inflamação/imunologia , Macrófagos Alveolares/imunologia , Tuberculose/imunologia , Animais , Citocinas/imunologia , Feminino , Inflamação/patologia , Macrófagos Alveolares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Tuberculose/patologia
5.
J Infect Dis ; 220(12): 1989-1998, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31412123

RESUMO

BACKGROUND: Tuberculosis is caused by Mycobacterium tuberculosis. Recent emergence of multidrug-resistant (MDR) tuberculosis strains seriously threatens tuberculosis control and prevention. However, the role of macrophage multidrug resistance gene MDR1 on intracellular M. tuberculosis survival during antituberculosis drug treatment is not known. METHODS: We used the human monocyte-derived macrophages to study the role of M. tuberculosis in regulation of MDR1 and drug resistance. RESULTS: We discovered that M. tuberculosis infection increases the expression of macrophage MDR1 to extrude various chemical substances, including tuberculosis drugs, resulting in enhanced survival of intracellular M. tuberculosis. The pathway of regulation involves M. tuberculosis infection of macrophages and suppression of heat shock factor 1, a transcriptional regulator of MDR1 through the up-regulation of miR-431. Notably, nonpathogenic Mycobacterium smegmatis did not increase MDR1 expression, indicating active secretion of virulence factors in pathogenic M. tuberculosis contributing to this phenotype. Finally, inhibition of MDR1 improves antibiotic-mediated killing of M. tuberculosis. CONCLUSION: We report a novel finding that M. tuberculosis up-regulates MDR1 during infection, which limits the exposure of M. tuberculosis to sublethal concentrations of antimicrobials. This condition promotes M. tuberculosis survival and potentially enhances the emergence of resistant variants.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Regulação da Expressão Gênica , Macrófagos/metabolismo , Macrófagos/microbiologia , Mycobacterium tuberculosis/fisiologia , Tuberculose/genética , Tuberculose/microbiologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Animais , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/imunologia , Camundongos , MicroRNAs/genética , Viabilidade Microbiana/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Tuberculose/tratamento farmacológico , Tuberculose/metabolismo , Tuberculose Pulmonar/genética , Tuberculose Pulmonar/metabolismo , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia , Fatores de Virulência
6.
J Infect Dis ; 220(3): 514-523, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-30923818

RESUMO

As we age, there is an increased risk for the development of tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) infection. Few studies consider that age-associated changes in the alveolar lining fluid (ALF) may increase susceptibility by altering soluble mediators of innate immunity. We assessed the impact of adult or elderly human ALF during Mtb infection in vitro and in vivo. We identified amplification of pro-oxidative and proinflammatory pathways in elderly ALF and decreased binding capability of surfactant-associated surfactant protein A (SP-A) and surfactant protein D (SP-D) to Mtb. Human macrophages infected with elderly ALF-exposed Mtb had reduced control and fewer phagosome-lysosome fusion events, which was reversed when elderly ALF was replenished with functional SP-A/SP-D. In vivo, exposure to elderly ALF exacerbated Mtb infection in young mice. Our studies demonstrate how the pulmonary environment changes as we age and suggest that Mtb may benefit from declining host defenses in the lung mucosa of the elderly.


Assuntos
Pulmão/imunologia , Pulmão/microbiologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia , Tuberculose/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Imunidade Inata/imunologia , Inflamação/imunologia , Inflamação/microbiologia , Lisossomos/imunologia , Lisossomos/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Pessoa de Meia-Idade , Mycobacterium tuberculosis/imunologia , Fagossomos/imunologia , Fagossomos/microbiologia , Proteína A Associada a Surfactante Pulmonar/imunologia , Proteína D Associada a Surfactante Pulmonar/imunologia , Tuberculose/microbiologia , Adulto Jovem
7.
J Immunol ; 198(5): 1985-1994, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28093527

RESUMO

Macrophage intracellular pathogen killing is defective in cystic fibrosis (CF), despite abundant production of reactive oxygen species (ROS) in lung tissue. Burkholderia species can cause serious infection in CF and themselves affect key oxidase components in murine non-CF cells. However, it is unknown whether human CF macrophages have an independent defect in the oxidative burst and whether Burkholderia contributes to this defect in terms of assembly of the NADPH oxidase complex and subsequent ROS production. In this article, we analyze CF and non-CF human monocyte-derived macrophages (MDMs) for ROS production, NADPH assembly capacity, protein kinase C expression, and calcium release in response to PMA and CF pathogens. CF MDMs demonstrate a nearly 60% reduction in superoxide production after PMA stimulation compared with non-CF MDMs. Although CF MDMs generally have increased total NADPH component protein expression, they demonstrate decreased expression of the calcium-dependent protein kinase C conventional subclass α/ß leading to reduced phosphorylation of NADPH oxidase components p47 phox and p40 phox in comparison with non-CF MDMs. Ingestion of B. cenocepacia independently contributes to and worsens the overall oxidative burst deficits in CF MDMs compared with non-CF MDMs. Together, these results provide evidence for inherent deficits in the CF macrophage oxidative burst caused by decreased phosphorylation of NADPH oxidase cytosolic components that are augmented by Burkholderia These findings implicate a critical role for defective macrophage oxidative responses in persistent bacterial infections in CF and create new opportunities for boosting the macrophage immune response to limit infection.


Assuntos
Infecções por Burkholderia/imunologia , Burkholderia cenocepacia/imunologia , Fibrose Cística/imunologia , Macrófagos/imunologia , NADPH Oxidases/metabolismo , Proteína Quinase C/metabolismo , Explosão Respiratória , Animais , Cálcio/metabolismo , Células Cultivadas , Regulação para Baixo , Humanos , Camundongos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo
8.
J Immunol ; 197(12): 4727-4735, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27913648

RESUMO

Mycobacterium tuberculosis imposes a large global health burden as the airborne agent of tuberculosis. Mycobacterium tuberculosis has been flourishing in human populations for millennia and is therefore highly adapted to the lung environment. Alveolar macrophages, a major host cell niche for M. tuberculosis, are not only phagocytose inhaled microbes and particulate matter but are also crucial in catabolizing lung surfactant, a lipid-protein complex that lines the alveolar spaces. Because macrophage host defense properties can be regulated by surfactant and M. tuberculosis can use host lipids as a carbon source during infection, we sought to determine the receptor(s) involved in surfactant lipid uptake by human macrophages and whether the presence of those lipids within macrophages prior to infection with M. tuberculosis enhances bacterial growth. We show that preformed scavenger receptor CD36 is redistributed to the cell membrane following exposure to surfactant lipids and surfactant protein A. Subsequently, surfactant lipids and/or surfactant protein A enhance CD36 transcript and protein levels. We show that CD36 participates in surfactant lipid uptake by human macrophages, as CD36 knockdown reduces uptake of dipalmitoylphosphatidylcholine, the most prevalent surfactant lipid species. Finally, exposing human macrophages to surfactant lipids prior to infection augments M. tuberculosis growth in a CD36-dependent manner. Thus, we provide evidence that CD36 mediates surfactant lipid uptake by human macrophages and that M. tuberculosis exploits this function for growth.


Assuntos
Antígenos CD36/metabolismo , Espaço Intracelular/microbiologia , Macrófagos Alveolares/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar/imunologia , 1,2-Dipalmitoilfosfatidilcolina/metabolismo , Antígenos CD36/genética , Células Cultivadas , Humanos , Imunidade Inata , Metabolismo dos Lipídeos , Pulmão/patologia , Macrófagos Alveolares/microbiologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Fagocitose , Proteína A Associada a Surfactante Pulmonar/metabolismo , Surfactantes Pulmonares/metabolismo , RNA Interferente Pequeno/genética
9.
J Immunol ; 197(6): 2390-9, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27534554

RESUMO

During Gram-negative bacterial infections, excessive LPS induces inflammation and sepsis via action on immune cells. However, the bulk of LPS can be cleared from circulation by the liver. Liver clearance is thought to be a slow process mediated exclusively by phagocytic resident macrophages, Kupffer cells (KC). However, we discovered that LPS disappears rapidly from the circulation, with a half-life of 2-4 min in mice, and liver eliminates about three quarters of LPS from blood circulation. Using microscopic techniques, we found that ∼75% of fluor-tagged LPS in liver became associated with liver sinusoidal endothelial cells (LSEC) and only ∼25% with KC. Notably, the ratio of LSEC-KC-associated LPS remained unchanged 45 min after infusion, indicating that LSEC independently processes the LPS. Most interestingly, results of kinetic analysis of LPS bioactivity, using modified limulus amebocyte lysate assay, suggest that recombinant factor C, an LPS binding protein, competitively inhibits high-density lipoprotein (HDL)-mediated LPS association with LSEC early in the process. Supporting the previous notion, 3 min postinfusion, 75% of infused fluorescently tagged LPS-HDL complex associates with LSEC, suggesting that HDL facilitates LPS clearance. These results lead us to propose a new paradigm of LSEC and HDL in clearing LPS with a potential to avoid inflammation during sepsis.


Assuntos
Células Endoteliais/fisiologia , Lipopolissacarídeos/sangue , Lipopolissacarídeos/metabolismo , Lipoproteínas HDL/metabolismo , Fígado/citologia , Proteínas de Fase Aguda/imunologia , Proteínas de Fase Aguda/metabolismo , Animais , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Células Endoteliais/imunologia , Infecções por Bactérias Gram-Negativas/imunologia , Meia-Vida , Inflamação/imunologia , Inflamação/prevenção & controle , Cinética , Células de Kupffer/imunologia , Lipopolissacarídeos/imunologia , Lipoproteínas HDL/imunologia , Fígado/imunologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Sepse/imunologia
10.
J Nanosci Nanotechnol ; 18(2): 925-935, 2018 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-29448516

RESUMO

Gallium doped mesoporous TiO2 with different weight percentages were synthesized by sol-gel method using Pluronic P123 as the structure directing template. The physico-chemical properties of all the synthesized catalysts were determined by XRD, TEM, SEM-EDAX, N2 adsorption-desorption studies, XPS, UV-vis DRS, FT-IR and photoluminescence spectroscopy. 1.0 wt% Ga-TiO2 exhibited the highest photocatalytic efficiency among all the synthesized materials under visible light due to the high surface area, reduced band gap and suppressed electron-hole recombination. Ga3+ ions substitutions for Ti4+ ions in TiO2 lattice created oxygen vacancies in TiO2 lattice, which created a defect energy level below the conduction band of TiO2 and hence the band gap was reduced. The oxygen vacancy defects was playing significant role to improve the adsorption of oxygen molecules, hydroxide ions and cationic rhodamine B (RhB) on TiO2 surface in an aqueous medium. The lifetime of the charge carriers was also enhanced by trapping the photogenerated electrons in oxygen vacancies and transferring them to the adsorbed O2 to produce superoxide anion radicals (O-. 2 ). The photo-induced holes at valence band reduced the adsorbed OH- ions and produced a large number of .OH radicals, which subsequently degraded the RhB. Hence oxygen vacancies created by gallium doping on TiO2 enhanced the photocatalytic efficiency for the degradation of RhB under visible light.

11.
Semin Immunol ; 26(6): 471-85, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25453226

RESUMO

Macrophages, the major host cells harboring Mycobacterium tuberculosis (M.tb), are a heterogeneous cell type depending on their tissue of origin and host they are derived from. Significant discord in macrophage responses to M.tb exists due to differences in M.tb strains and the various types of macrophages used to study tuberculosis (TB). This review will summarize current concepts regarding macrophage responses to M.tb infection, while pointing out relevant differences in experimental outcomes due to the use of divergent model systems. A brief description of the lung environment is included since there is increasing evidence that the alveolar macrophage (AM) has immunoregulatory properties that can delay optimal protective host immune responses. In this context, this review focuses on selected macrophage immunoregulatory pattern recognition receptors (PRRs), cytokines, negative regulators of inflammation, lipid mediators and microRNAs (miRNAs).


Assuntos
Pulmão/imunologia , Macrófagos Alveolares/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar/imunologia , Citocinas/genética , Citocinas/imunologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Pulmão/microbiologia , Pulmão/patologia , Ativação de Macrófagos , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/patologia , MicroRNAs/genética , MicroRNAs/imunologia , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/imunologia , Tuberculose Pulmonar/genética , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia
12.
J Immunol ; 195(5): 2019-29, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26202986

RESUMO

γ-Tilmanocept ((99m)Tc-labeled-tilmanocept or [(99m)Tc]-tilmanocept) is the first mannose-containing, receptor-directed, radiolabeled tracer for the highly sensitive imaging of sentinel lymph nodes in solid tumor staging. To elucidate the mannose-binding receptor that retains tilmanocept in this microenvironment, human macrophages were used that have high expression of the C-type lectin mannose receptor (MR; CD206). Cy3-labeled tilmanocept exhibited high specificity binding to macrophages that was nearly abolished in competitive inhibition experiments. Furthermore, Cy3-tilmanocept binding was markedly reduced on macrophages deficient in the MR by small interfering RNA treatment and was increased on MR-transfected HEK 293 cells. Finally, confocal microscopy revealed colocalization of Cy3-tilmanocept with the macrophage membrane MR and binding of labeled tilmanocept to MR(+) cells (macrophages and/or dendritic cells) in human sentinel lymph node tissues. Together these data provide strong evidence that CD206 is a major binding receptor for γ-tilmanocept. Identification of CD206 as the γ-tilmanocept-binding receptor enables opportunities for designing receptor-targeted advanced imaging agents and therapeutics for cancer and other diseases.


Assuntos
Dextranos/metabolismo , Lectinas Tipo C/metabolismo , Mananas/metabolismo , Lectinas de Ligação a Manose/metabolismo , Receptores de Superfície Celular/metabolismo , Biópsia de Linfonodo Sentinela/métodos , Pentetato de Tecnécio Tc 99m/análogos & derivados , Carbocianinas/química , Células Cultivadas , Dextranos/química , Citometria de Fluxo , Células HEK293 , Humanos , Imuno-Histoquímica , Lectinas Tipo C/genética , Linfonodos/metabolismo , Macrófagos/metabolismo , Mananas/química , Receptor de Manose , Lectinas de Ligação a Manose/genética , Microscopia Confocal , Estrutura Molecular , Neoplasias/diagnóstico , Neoplasias/metabolismo , Ligação Proteica , Interferência de RNA , Compostos Radiofarmacêuticos/metabolismo , Receptores de Superfície Celular/genética , Pentetato de Tecnécio Tc 99m/química , Pentetato de Tecnécio Tc 99m/metabolismo
13.
J Immunol ; 193(9): 4537-47, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25252958

RESUMO

IFN-γ-activated macrophages play an essential role in controlling intracellular pathogens; however, macrophages also serve as the cellular home for the intracellular pathogen Mycobacterium tuberculosis. Based on previous evidence that M. tuberculosis can modulate host microRNA (miRNA) expression, we examined the miRNA expression profile of M. tuberculosis-infected primary human macrophages. We identified 31 differentially expressed miRNAs in primary human macrophages during M. tuberculosis infection by NanoString and confirmed our findings by quantitative real-time RT-PCR. In addition, we determined a role for two miRNAs upregulated upon M. tuberculosis infection, miR-132 and miR-26a, as negative regulators of transcriptional coactivator p300, a component of the IFN-γ signaling cascade. Knockdown expression of miR-132 and miR-26a increased p300 protein levels and improved transcriptional, translational, and functional responses to IFN-γ in human macrophages. Collectively, these data validate p300 as a target of miR-132 and miR-26a, and demonstrate a mechanism by which M. tuberculosis can limit macrophage responses to IFN-γ by altering host miRNA expression.


Assuntos
Macrófagos/imunologia , Macrófagos/metabolismo , MicroRNAs/genética , Mycobacterium tuberculosis/imunologia , Tuberculose/genética , Tuberculose/imunologia , Regiões 3' não Traduzidas , Sítios de Ligação , Linhagem Celular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Antígenos HLA-DR/genética , Antígenos HLA-DR/imunologia , Humanos , Interferon gama/farmacologia , Macrófagos/efeitos dos fármacos , Interferência de RNA , RNA Mensageiro/genética , Receptores de IgG/genética , Receptores de IgG/metabolismo , Reprodutibilidade dos Testes , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
14.
PLoS Pathog ; 9(1): e1003114, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23359218

RESUMO

Complement receptor 3 (CR3, CD11b/CD18) is a major macrophage phagocytic receptor. The biochemical pathways through which CR3 regulates immunologic responses have not been fully characterized. Francisella tularensis is a remarkably infectious, facultative intracellular pathogen of macrophages that causes tularemia. Early evasion of the host immune response contributes to the virulence of F. tularensis and CR3 is an important receptor for its phagocytosis. Here we confirm that efficient attachment and uptake of the highly virulent Type A F. tularensis spp. tularensis strain Schu S4 by human monocyte-derived macrophages (hMDMs) requires complement C3 opsonization and CR3. However, despite a>40-fold increase in uptake following C3 opsonization, Schu S4 induces limited pro-inflammatory cytokine production compared with non-opsonized Schu S4 and the low virulent F. novicida. This suggests that engagement of CR3 by opsonized Schu S4 contributes specifically to the immune suppression during and shortly following phagocytosis which we demonstrate by CD11b siRNA knockdown in hMDMs. This immune suppression is concomitant with early inhibition of ERK1/2, p38 MAPK and NF-κB activation. Furthermore, TLR2 siRNA knockdown shows that pro-inflammatory cytokine production and MAPK activation in response to non-opsonized Schu S4 depends on TLR2 signaling providing evidence that CR3-TLR2 crosstalk mediates immune suppression for opsonized Schu S4. Deletion of the CD11b cytoplasmic tail reverses the CR3-mediated decrease in ERK and p38 activation during opsonized Schu-S4 infection. The CR3-mediated signaling pathway involved in this immune suppression includes Lyn kinase and Akt activation, and increased MKP-1, which limits TLR2-mediated pro-inflammatory responses. These data indicate that while the highly virulent F. tularensis uses CR3 for efficient uptake, optimal engagement of this receptor down-regulates TLR2-dependent pro-inflammatory responses by inhibiting MAPK activation through outside-in signaling. CR3-linked immune suppression is an important mechanism involved in the pathogenesis of F. tularensis infection.


Assuntos
Francisella tularensis/imunologia , Tolerância Imunológica , Inflamação/imunologia , Antígeno de Macrófago 1/metabolismo , Fagocitose/imunologia , Tularemia/imunologia , Animais , Células CHO/imunologia , Células CHO/metabolismo , Cricetinae , Cricetulus , Inativação Gênica , Humanos , Evasão da Resposta Imune , Fatores Imunológicos/metabolismo , Inflamação/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas Opsonizantes/metabolismo , RNA Interferente Pequeno/genética , Transfecção , Tularemia/metabolismo
15.
Br J Biomed Sci ; 72(3): 102-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26510264

RESUMO

Mitral stenosis (MS) causes stagnation of blood flow, leading to thrombus formation in the left atrium (LA), which may lead to systemic thrombo-embolic complications and stroke. We compared the alterations in echocardiographic and procoagulant parameters in patients with severe rheumatic MS with and without LA thrombus. The study was a cross-sectional study of patients with rheumatic MS, being evaluated for percutaneous mitral commisurotomy. Group 1 patients comprised of patients with rheumatic MS with LA thrombus (n=35) and Group 2 patients had rheumatic MS without LA thrombus (n = 45). Platelet aggregability, fibrinogen, homocysteine, vitamin B12 and folate; mitral valve area (MVA), mean mitral gradient and pulmonary artery pressure (PAP) were assessed in all study subjects. Significant increase in fibrinogen, homocysteine and platelet aggregation and fall in homocysteine-associated determinants were seen in Group 1, as compared with Group 2. Raised fibrinogen, lowered homocysteine-vitamin determinants and lowered mitral valve area were associated independently, with presence of LA thrombus in rheumatic MS. In this study, fibrinogen, vitamin B12 and folate were independently associated with the occurrence of thrombus in patients with rheumatic MS. Hence, our results suggest that increase in procoagulant mechanisms contribute to increased risk of thrombosis in the left atrium in patients with rheumatic MS.


Assuntos
Fibrinogênio/metabolismo , Ácido Fólico/sangue , Homocisteína/metabolismo , Estenose da Valva Mitral/sangue , Cardiopatia Reumática/sangue , Trombose/sangue , Vitamina B 12/sangue , Adulto , Biomarcadores/sangue , Plaquetas/metabolismo , Plaquetas/patologia , Estudos Transversais , Feminino , Átrios do Coração/diagnóstico por imagem , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Valva Mitral/diagnóstico por imagem , Valva Mitral/metabolismo , Valva Mitral/patologia , Estenose da Valva Mitral/complicações , Estenose da Valva Mitral/diagnóstico por imagem , Estenose da Valva Mitral/patologia , Agregação Plaquetária , Cardiopatia Reumática/complicações , Cardiopatia Reumática/diagnóstico por imagem , Cardiopatia Reumática/patologia , Trombose/complicações , Trombose/diagnóstico por imagem , Trombose/patologia , Ultrassonografia
16.
J Infect Dis ; 209(7): 1045-54, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24265438

RESUMO

Mycobacteria produce an unusual, glycolylated form of muramyl dipeptide (MDP) that is more potent and efficacious at inducing NOD2-mediated host responses. We tested the importance of this modified form of MDP in Mycobacterium tuberculosis by disrupting the gene, namH, responsible for this modification. In vitro, the namH mutant did not produce N-glycolylated muropeptides, but there was no alteration in colony morphology, growth kinetics, cellular morphology, or mycolic acid profile. Ex vivo, the namH mutant survived and replicated normally in murine and human macrophages, yet induced diminished production of tumor necrosis factor α. In vivo, namH disruption did not affect the bacterial burden during infection of C57BL/6 mice or cellular recruitment to the lungs but modestly prolonged survival after infection in Rag1(-/-) mice. These results indicate that the modified MDP is an important contributor to the unusual immunogenicity of mycobacteria but has a limited role in the pathogenesis of M. tuberculosis infection.


Assuntos
Acetilmuramil-Alanil-Isoglutamina/imunologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Peptidoglicano/imunologia , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Animais , Carga Bacteriana , Células Cultivadas , Modelos Animais de Doenças , Deleção de Genes , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/enzimologia , Peptidoglicano/química , Processamento de Proteína Pós-Traducional , Análise de Sobrevida , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Virulência
17.
Clin Immunol ; 151(1): 66-77, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24525049

RESUMO

Females of child-bearing age are more resistant to infectious disease and have an increased risk of systemic lupus erythematosus (SLE). We hypothesized that estrogen-induced gene expression could establish an immunoactivated state which would render enhanced defense against infection, but may be deleterious in autoimmune development. Using peripheral blood mononuclear cells (PBMCs), we demonstrate enhanced responses with immunogen stimulation in the presence of 17ß-estradiol (E2) and gene array analyses reveal toll-like receptor 8 (TLR8) as an E2-responsive candidate gene. TLR8 expression levels are up-regulated in SLE and PBMCs stimulated with TLR8 agonist display a female sex-biased, E2-sensitive response. Moreover, we identify a putative ERα-binding region near the TLR8 locus and blocking ERα expression significantly decreases E2-mediated TLR8 induction. Our findings characterize TLR8 as a novel estrogen target gene that can lower the inflammatory threshold and implicate an IFNα-independent inflammatory mechanism that could contribute to higher SLE incidence in women.


Assuntos
Endossomos/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Lúpus Eritematoso Sistêmico/imunologia , Receptor 8 Toll-Like/imunologia , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Células Cultivadas , Endossomos/imunologia , Endossomos/metabolismo , Receptor alfa de Estrogênio/genética , Feminino , Regulação da Expressão Gênica , Humanos , Imidazóis/farmacologia , Fatores Imunológicos/farmacologia , Interferon-alfa/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Fatores Sexuais , Transdução de Sinais , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/genética
18.
J Nanosci Nanotechnol ; 14(4): 3187-92, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24734753

RESUMO

Single-site CeAlPO-5 with Al/Ce ratios 25, 50, 75, 100 and 125 were synthesized hydrothermally in fluoride medium. The XRD patterns of CeAlPO-5 exhibited characteristic reflections of AlPO-5. 27Al MAS-NMR of CeAIPO-5(25) showed two unusual peaks at -20.78 and -71.35 ppm due to delocalization of cerium unpaired electron. However, 31P MAS-NMR exhibited the usual characteristic peak similar to that of AlPO-5. Vapour phase oxidation of toluene in air over CeAlPO-5 yielded benzaldehyde with high toluene conversion. The time on stream study established the stability of the catalyst. This catalyst can also be used for the selective oxidation of other alkyl aromatics.

19.
Proc Natl Acad Sci U S A ; 108(42): 17408-13, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-21969554

RESUMO

Contact of Mycobacterium tuberculosis (M.tb) with the immune system requires interactions between microbial surface molecules and host pattern recognition receptors. Major M.tb-exposed cell envelope molecules, such as lipomannan (LM), contain subtle structural variations that affect the nature of the immune response. Here we show that LM from virulent M.tb (TB-LM), but not from avirulent Myocobacterium smegmatis (SmegLM), is a potent inhibitor of TNF biosynthesis in human macrophages. This difference in response is not because of variation in Toll-like receptor 2-dependent activation of the signaling kinase MAPK p38. Rather, TB-LM stimulation leads to destabilization of TNF mRNA transcripts and subsequent failure to produce TNF protein. In contrast, SmegLM enhances MAPK-activated protein kinase 2 phosphorylation, which is critical for maintaining TNF mRNA stability in part by contributing microRNAs (miRNAs). In this context, human miRNA miR-125b binds to the 3' UTR region of TNF mRNA and destabilizes the transcript, whereas miR-155 enhances TNF production by increasing TNF mRNA half-life and limiting expression of SHIP1, a negative regulator of the PI3K/Akt pathway. We show that macrophages incubated with TB-LM and live M.tb induce high miR-125b expression and low miR-155 expression with correspondingly low TNF production. In contrast, SmegLM and live M. smegmatis induce high miR-155 expression and low miR-125b expression with high TNF production. Thus, we identify a unique cellular mechanism underlying the ability of a major M.tb cell wall component, TB-LM, to block TNF biosynthesis in human macrophages, thereby allowing M.tb to subvert host immunity and potentially increase its virulence.


Assuntos
Lipopolissacarídeos/imunologia , MAP Quinase Quinase 2/metabolismo , Macrófagos/imunologia , MicroRNAs/metabolismo , Mycobacterium tuberculosis/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Humanos , Técnicas In Vitro , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/microbiologia , MicroRNAs/genética , Mycobacterium tuberculosis/patogenicidade , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estabilidade de RNA , Receptor 2 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/genética , Virulência/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Proteomics ; 13(14): 2113-24, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23595970

RESUMO

The current treatment for glioblastoma includes temozolomide (TMZ) chemotherapy, yet the mechanism of action of TMZ is not thoroughly understood. Here, we investigated the TMZ-induced changes in the proteome of the glioma-derived cell line (U251) by 2D DIGE. We found 95 protein spots to be significantly altered in their expression after TMZ treatment. MS identified four upregulated spots: aspartyl tRNA synthetase glutathione synthetase, interleukin-1 receptor-associated kinase-4 (IRAK4), and breast carcinoma amplified sequence-1 and one downregulated spot: optineurin. TMZ-induced regulation of these five genes was validated by RT-qPCR and Western blot analysis. RNAi-mediated knockdown of IRAK4, an important mediator of Toll-like receptors signaling and chemoresistance, rendered the glioma cells resistant to TMZ. High levels of IRAK4 induced upon TMZ treatment resulted in IRAK1 downregulation and inhibition of NFkB pathway. Endogenous IRAK4 protein, but not transcript levels in glioma cell lines, correlated with TMZ sensitivity. Thus, we have identified several TMZ-modulated proteins and discovered an important novel role for IRAK4 in determining TMZ sensitivity of glioma cells through its ability to inhibit Toll-like receptor signaling and NFkB pathway.


Assuntos
Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioma/tratamento farmacológico , Glioma/metabolismo , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Proteoma/efeitos dos fármacos , Antineoplásicos Alquilantes/farmacologia , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Análise por Conglomerados , Dacarbazina/farmacologia , Dimetil Sulfóxido , Eletroforese em Gel Bidimensional , Humanos , Quinases Associadas a Receptores de Interleucina-1/análise , Quinases Associadas a Receptores de Interleucina-1/genética , NF-kappa B/metabolismo , Proteoma/análise , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Temozolomida , Receptores Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA