Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Immunity ; 47(6): 1169-1181.e7, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29246444

RESUMO

The tumor suppressor PTEN controls cell proliferation by regulating phosphatidylinositol-3-kinase (PI3K) activity, but the participation of PTEN in host defense against bacterial infection is less well understood. Anti-inflammatory PI3K-Akt signaling is suppressed in patients with cystic fibrosis (CF), a disease characterized by hyper-inflammatory responses to airway infection. We found that Ptenl-/- mice, which lack the NH2-amino terminal splice variant of PTEN, were unable to eradicate Pseudomonas aeruginosa from the airways and could not generate sufficient anti-inflammatory PI3K activity, similar to what is observed in CF. PTEN and the CF transmembrane conductance regulator (CFTR) interacted directly and this interaction was necessary to position PTEN at the membrane. CF patients under corrector-potentiator therapy, which enhances CFTR transport to the membrane, have increased PTEN amounts. These findings suggest that improved CFTR trafficking could enhance P. aeruginosa clearance from the CF airway by activating PTEN-mediated anti-bacterial responses and might represent a therapeutic strategy.


Assuntos
Membrana Celular/imunologia , Regulador de Condutância Transmembrana em Fibrose Cística/imunologia , Fibrose Cística/imunologia , PTEN Fosfo-Hidrolase/imunologia , Infecções por Pseudomonas/imunologia , Aminofenóis/farmacologia , Aminopiridinas/farmacologia , Animais , Benzodioxóis/farmacologia , Membrana Celular/efeitos dos fármacos , Fibrose Cística/tratamento farmacológico , Fibrose Cística/genética , Fibrose Cística/microbiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/microbiologia , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Ligação Proteica , Conformação Proteica , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/imunologia , Quinolonas/farmacologia , Transdução de Sinais
2.
Artigo em Inglês | MEDLINE | ID: mdl-38656811

RESUMO

Pseudomonas aeruginosa causes chronic lung infection in cystic fibrosis (CF), resulting in structural lung damage and progressive pulmonary decline. P. aeruginosa in the CF lung undergoes numerous changes, adapting to host-specific airway pressures while establishing chronic infection. P. aeruginosa undergoes lipid A structural modification during CF chronic infection, not seen in any other disease state. Lipid A, the membrane anchor of lipopolysaccharide (i.e., endotoxin), comprises the majority of the outer membrane of Gram-negative bacteria and is a potent toll-like receptor (TLR)4 agonist. The structure of P. aeruginosa lipid A is intimately linked with its recognition by TLR4, and subsequent immune response. Prior work has identified P. aeruginosa strains with altered lipid A structures that arise during chronic CF lung infection; however, the impact of P. aeruginosa lipid A structure on airway disease has not been investigated. Here, we show that P. aeruginosa lipid A lacks PagL-mediated deacylation during human airway infection using a direct-from-sample mass spectrometry approach on human bronchoalveolar lavage fluid. This structure triggers increased pro-inflammatory cytokine production by primary human macrophages. Furthermore, alterations in lipid A 2-hydroxylation impact cytokine response in a site-specific manner, independent of CFTR function. Interestingly, there is a CF-specific reduction in IL-8 secretion within the epithelial-cell compartment that only occurs in CF bronchial epithelial cells when infected with CF-adapted P. aeruginosa that lack PagL-mediated lipid A deacylation. Taken together, we show that P. aeruginosa alters its lipid A structure during acute lung infection and that this lipid A structure induces stronger signaling through TLR4.

3.
Respir Res ; 21(1): 326, 2020 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-33302964

RESUMO

Pulmonary infections are associated with a brisk inflammatory reaction to bacterial surface components. Lipopolysaccharides (LPS) trigger macrophage activation and release of mitochondrial metabolites that control the intensity of the immune response. Whereas succinate induces oxidative stress (ROS), HIF1α stabilization, glycolysis and IL-1ß release, itaconate suppresses inflammation by inhibiting succinate oxidation, glycolytic flux and promoting anti-oxidant Nrf2-HO-1 functions. P. aeruginosa is a major pathogen associated with acute and chronic lung infection. Although both secreted toxins, LPS and proteases are key factors to establish acute P. aeruginosa pneumonia, lack of these components in chronic P. aeruginosa isolates suggest these organisms exploit other mechanisms to adapt and persist in the lung. Upon inhalation, P. aeruginosa strains trigger airway macrophage reprograming and bacterial variants obtained from acutely and chronically infected subjects exhibit metabolic adaptation consistent with succinate and itaconate assimilation; namely, high expression of extracellular polysaccharides (EPS), reduced lptD-LPS function, increased glyoxylate shunt (GS) activity and substantial biofilm production. In this review we discuss recent findings illustrating how P. aeruginosa induces and adapts to macrophage metabolites in the human lung, and that catabolism of succinate and itaconate contribute to their formidable abilities to tolerate oxidative stress, phagocytosis and immune clearance.


Assuntos
Metabolismo Energético , Pulmão/microbiologia , Ativação de Macrófagos , Macrófagos Alveolares/microbiologia , Pneumonia Bacteriana/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Animais , Biofilmes/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Estresse Oxidativo , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/metabolismo , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/imunologia , Espécies Reativas de Oxigênio/metabolismo
4.
Nature ; 512(7514): 295-8, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25119028

RESUMO

The seismic gap theory identifies regions of elevated hazard based on a lack of recent seismicity in comparison with other portions of a fault. It has successfully explained past earthquakes (see, for example, ref. 2) and is useful for qualitatively describing where large earthquakes might occur. A large earthquake had been expected in the subduction zone adjacent to northern Chile, which had not ruptured in a megathrust earthquake since a M âˆ¼8.8 event in 1877. On 1 April 2014 a M 8.2 earthquake occurred within this seismic gap. Here we present an assessment of the seismotectonics of the March-April 2014 Iquique sequence, including analyses of earthquake relocations, moment tensors, finite fault models, moment deficit calculations and cumulative Coulomb stress transfer. This ensemble of information allows us to place the sequence within the context of regional seismicity and to identify areas of remaining and/or elevated hazard. Our results constrain the size and spatial extent of rupture, and indicate that this was not the earthquake that had been anticipated. Significant sections of the northern Chile subduction zone have not ruptured in almost 150 years, so it is likely that future megathrust earthquakes will occur to the south and potentially to the north of the 2014 Iquique sequence.

5.
Am J Respir Cell Mol Biol ; 60(2): 158-166, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30183325

RESUMO

IFN-λ and IL-22, cytokines that share the coreceptor IL-10RB, are both induced over the course of Klebsiella pneumoniae ST258 (KP35) pneumonia. IL-22 is known to protect mucosal barriers, whereas the effects of IFN-λ on the mucosa are not established. We postulated that IFN-λ plays a role in regulating the airway epithelial barrier to facilitate cellular trafficking to the site of infection. In response to IFN-λ, the transmigration of neutrophils across a polarized monolayer of airway epithelial cells was increased, consistent with diminished epithelial integrity. KP35 infection increased epithelial permeability, and pretreatment with IFN-λ amplified this effect and facilitated bacterial transmigration. These effects of IFN-λ were confirmed in vivo, in that mice lacking the receptor for IFN-λ (Ifnlr1-/-) were protected from bacteremia in a murine model of KP35 pneumonia. Conversely, the integrity of the epithelial barrier was protected by IL-22, with subsequent impairment of neutrophil and bacterial transmigration in vitro. Maximal expression of IL-22 in vivo was observed later in the course of infection than IFN-λ production, with high levels of IL-22 produced by recruited immune cells at 48 hours, consistent with a role in epithelial barrier recovery. The divergent and opposing expression of these two related cytokines suggests a regulated interaction in the host response to KP35 infection. A major physiological effect of IFN-λ signaling is a decrease in epithelial barrier integrity, which facilitates immune cell recruitment but also enables K. pneumoniae invasion.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Interferons/metabolismo , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/patogenicidade , Animais , Bacteriemia/genética , Bacteriemia/microbiologia , Brônquios/citologia , Líquido da Lavagem Broncoalveolar/microbiologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Humanos , Interferons/farmacologia , Subunidade beta de Receptor de Interleucina-10/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Interleucinas/farmacologia , Klebsiella pneumoniae/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neutrófilos/microbiologia , Neutrófilos/patologia , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Interleucina 22
6.
Am J Respir Cell Mol Biol ; 61(2): 185-197, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30742488

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA) is a versatile human pathogen that is associated with diverse types of infections ranging from benign colonization to sepsis. We postulated that MRSA must undergo specific genotypic and phenotypic changes to cause chronic pulmonary disease. We investigated how MRSA adapts to the human airway to establish chronic infection, as occurs during cystic fibrosis (CF). MRSA isolates from patients with CF that were collected over a 4-year period were analyzed by whole-genome sequencing, transcriptional analysis, and metabolic studies. Persistent MRSA infection was associated with staphylococcal metabolic adaptation, but not changes in immunogenicity. Adaptation was characterized by selective use of the tricarboxylic acid cycle cycle and generation of biofilm, a means of limiting oxidant stress. Increased transcription of specific metabolic genes was conserved in all host-adapted strains, most notably a 10,000-fold increase in fumC, which catalyzes the interconversion of fumarate and malate. Elevated fumarate levels promoted in vitro biofilm production in clinical isolates. Host-adapted strains preferred to assimilate glucose polymers and pyruvate, which can be metabolized to generate N-acetylglucosamine polymers that comprise biofilm. MRSA undergoes substantial metabolic adaptation to the human airway to cause chronic pulmonary infection, and selected metabolites may be useful therapeutically to inhibit infection.


Assuntos
Fibrose Cística/microbiologia , Pneumopatias/microbiologia , Staphylococcus aureus Resistente à Meticilina/metabolismo , Pneumonia Estafilocócica/microbiologia , Infecções Estafilocócicas/microbiologia , Acetilglucosamina/metabolismo , Adulto , Animais , Biofilmes , Brônquios/metabolismo , Líquido da Lavagem Broncoalveolar , Fibrose Cística/metabolismo , Citocinas/metabolismo , Feminino , Fumaratos/metabolismo , Gentamicinas/farmacologia , Glucose/metabolismo , Humanos , Pneumopatias/metabolismo , Malatos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Filogenia , Pneumonia Estafilocócica/metabolismo , Ácido Pirúvico/metabolismo , Infecções Estafilocócicas/metabolismo , Transcrição Gênica , Ácidos Tricarboxílicos/metabolismo , Sequenciamento Completo do Genoma
7.
J Immunol ; 199(1): 212-223, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28566367

RESUMO

Human respiratory syncytial virus (hRSV) is the leading cause of severe lower respiratory tract infections in children. The development of novel prophylactic and therapeutic antiviral drugs against hRSV is imperative to control the burden of disease in the susceptible population. In this study, we examined the effects of inducing the activity of the host enzyme heme oxygenase-1 (HO-1) on hRSV replication and pathogenesis on lung inflammation induced by this virus. Our results show that after hRSV infection, HO-1 induction with metalloporphyrin cobalt protoporphyrin IX significantly reduces the loss of body weight due to hRSV-induced disease. Further, HO-1 induction also decreased viral replication and lung inflammation, as evidenced by a reduced neutrophil infiltration into the airways, with diminished cytokine and chemokine production and reduced T cell function. Concomitantly, upon cobalt protoporphyrin IX treatment, there is a significant upregulation in the production of IFN-α/ß mRNAs in the lungs. Furthermore, similar antiviral and protective effects occur by inducing the expression of human HO-1 in MHC class II+ cells in transgenic mice. Finally, in vitro data suggest that HO-1 induction can modulate the susceptibility of cells, especially the airway epithelial cells, to hRSV infection.


Assuntos
Heme Oxigenase-1/metabolismo , Pulmão/imunologia , Infecções por Vírus Respiratório Sincicial/fisiopatologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/fisiologia , Animais , Linhagem Celular , Citocinas/biossíntese , Citocinas/imunologia , Replicação do DNA , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Heme Oxigenase-1/genética , Humanos , Interferon-alfa/biossíntese , Interferon-alfa/imunologia , Interferon beta/imunologia , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Camundongos , Protoporfirinas/administração & dosagem , Protoporfirinas/farmacologia , Infecções por Vírus Respiratório Sincicial/imunologia , Linfócitos T/imunologia , Ligação Viral , Internalização do Vírus , Replicação Viral
8.
J Plant Res ; 132(4): 509-520, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31250145

RESUMO

The expression of plant secondary metabolism is strongly controlled by plant both in time and space. Although the variation of secondary metabolites, such as soluble and structural phenolics (e.g., lignins), has been largely observed in gall-inducing insects, and compared to their non-galled host organs, only a few datasets recording such variation are available. Accordingly, the relative importance of spatiotemporal variability in phenolic contents, and the influence of gall developmental stages on the original composition of host organs are poorly discussed. To address this knowledge gap, we histochemically determined the sites of polyphenol and lignin accumulation, and the polyphenol contents in three developmental stages of two calophyid galls and their correspondent host organs. Current results indicate that the compartmentalization of phenolics and lignins on Schinus polygama (Cav.) Cabrera follows a similar pattern in the two-calophyid galls, accumulating in the outer (the external tissue layers) and in the inner tissue compartments (the cell layers in contact with the gall chamber). The non-accumulation in the median compartment (median parenchyma layers of gall wall with vascular bundles, where gall inducer feeds) is important for the inducer, because its mouth apparatus enter in contact with the cells of this compartment. Also, the concentration of phenolics has opposite dynamics, decreasing in leaf galls and increasing in stem galls, in temporal scale, i.e., from maturation toward senescence. The concentration of phenolics in non-galled host organs, and in both galls indicated the extended phenotype of Calophya rubra (Blanchard) and C. mammifex Burckhardt & Basset (Hemiptera: Sternorrhyncha: Psylloidea: Calophyidae) over the same host plant metabolic potentiality.


Assuntos
Anacardiaceae/parasitologia , Hemípteros/parasitologia , Fenóis/metabolismo , Tumores de Planta/parasitologia , Anacardiaceae/metabolismo , Animais , Lignina/metabolismo , Polifenóis/metabolismo
9.
Molecules ; 24(20)2019 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-31635434

RESUMO

Grape canes (Vitis vinifera L.) are a viticulture industry by-product with an important content of secondary metabolites, mainly polyphenols with a broad spectrum of demonstrated health benefits. Grape canes, therefore, have considerable economic potential as a source of high-value phytochemicals. In this work, liquid chromatography coupled with electrospray ionization hybrid linear trap quadrupole-Orbitrap mass spectrometry (LC-LTQ-Orbitrap) was used for the comprehensive identification of polyphenolic compounds in grape canes. Identification of polyphenols was performed by comparing their retention times, accurate mass measured, and mass fragmentation patterns with those of reference substances or available data in the literature. A total of 75 compounds were identified, including phenolic acids, flavanols, flavonols, flavanonols, flavanones, and stilbenoids. The most abundant polyphenols were proanthocyanidins and stilbenoids and their oligomers. Moreover, the high-resolution mass spectrometry analysis revealed the occurrence of 17 polyphenols never described before in grape canes, thereby providing a more complete polyphenolic profile of this potentially valuable by-product.


Assuntos
Polifenóis/química , Polifenóis/isolamento & purificação , Vitis/química , Cromatografia Líquida/métodos , Estrutura Molecular , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Espectrometria de Massas por Ionização por Electrospray/métodos
10.
Proc Natl Acad Sci U S A ; 111(31): E3214-23, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25056968

RESUMO

Human respiratory syncytial virus (hRSV) is the leading cause of bronchiolitis and pneumonia in young children worldwide. The recurrent hRSV outbreaks and reinfections are the cause of a significant public health burden and associate with an inefficient antiviral immunity, even after disease resolution. Although several mouse- and human cell-based studies have shown that hRSV infection prevents naïve T-cell activation by antigen-presenting cells, the mechanism underlying such inhibition remains unknown. Here, we show that the hRSV nucleoprotein (N) could be at least partially responsible for inhibiting T-cell activation during infection by this virus. Early after infection, the N protein was expressed on the surface of epithelial and dendritic cells, after interacting with trans-Golgi and lysosomal compartments. Further, experiments on supported lipid bilayers loaded with peptide-MHC (pMHC) complexes showed that surface-anchored N protein prevented immunological synapse assembly by naive CD4(+) T cells and, to a lesser extent, by antigen-experienced T-cell blasts. Synapse assembly inhibition was in part due to reduced T-cell receptor (TCR) signaling and pMHC clustering at the T-cell-bilayer interface, suggesting that N protein interferes with pMHC-TCR interactions. Moreover, N protein colocalized with the TCR independently of pMHC, consistent with a possible interaction with TCR complex components. Based on these data, we conclude that hRSV N protein expression at the surface of infected cells inhibits T-cell activation. Our study defines this protein as a major virulence factor that contributes to impairing acquired immunity and enhances susceptibility to reinfection by hRSV.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Membrana Celular/metabolismo , Sinapses Imunológicas/imunologia , Nucleoproteínas/metabolismo , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais/metabolismo , Animais , Brefeldina A/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/patologia , Comunicação Celular , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Antígenos de Histocompatibilidade/imunologia , Humanos , Sinapses Imunológicas/efeitos dos fármacos , Bicamadas Lipídicas/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/imunologia , Transporte Proteico/efeitos dos fármacos , Receptores de Antígenos de Linfócitos T/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Replicação Viral/efeitos dos fármacos
11.
Immunology ; 147(1): 55-72, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26451966

RESUMO

Human respiratory syncytial virus (hRSV) is the leading cause of infant hospitalization related to respiratory disease. Infection with hRSV produces abundant infiltration of immune cells into the airways, which combined with an exacerbated pro-inflammatory immune response can lead to significant damage to the lungs. Human RSV re-infection is extremely frequent, suggesting that this virus may have evolved molecular mechanisms that interfere with host adaptive immunity. Infection with hRSV can be reduced by administering a humanized neutralizing antibody against the virus fusion protein in high-risk infants. Although neutralizing antibodies against hRSV effectively block the infection of airway epithelial cells, here we show that both, bone marrow-derived dendritic cells (DCs) and lung DCs undergo infection with IgG-coated virus (hRSV-IC), albeit abortive. Yet, this is enough to negatively modulate DC function. We observed that such a process is mediated by Fcγ receptors (FcγRs) expressed on the surface of DCs. Remarkably, we also observed that in the absence of hRSV-specific antibodies FcγRIII knockout mice displayed significantly less cellular infiltration in the lungs after hRSV infection, compared with wild-type mice, suggesting a potentially harmful, IgG-independent role for this receptor in hRSV disease. Our findings support the notion that FcγRs can contribute significantly to the modulation of DC function by hRSV and hRSV-IC. Further, we provide evidence for an involvement of FcγRIII in the development of hRSV pathogenesis.


Assuntos
Células Dendríticas/metabolismo , Pulmão/metabolismo , Ativação Linfocitária , Receptores de IgG/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sincicial Respiratório Humano/patogenicidade , Linfócitos T/metabolismo , Imunidade Adaptativa , Animais , Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/metabolismo , Antivirais/farmacologia , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/virologia , Modelos Animais de Doenças , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/virologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Palivizumab/farmacologia , Receptores de IgG/deficiência , Receptores de IgG/genética , Receptores de IgG/imunologia , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/genética , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/imunologia , Transdução de Sinais , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/virologia , Carga Viral , Replicação Viral
12.
Immunology ; 149(1): 1-12, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26938875

RESUMO

Haem-oxygenase-1 (HO-1) is an enzyme responsible for the degradation of haem that can suppress inflammation, through the production of carbon monoxide (CO). It has been shown in several experimental models that genetic and pharmacological induction of HO-1, as well as non-toxic administration of CO, can reduce inflammatory diseases, such as endotoxic shock, type 1 diabetes and graft rejection. Recently, it was shown that the HO-1/CO system can alter the function of antigen-presenting cells (APCs) and reduce T-cell priming, which can be beneficial during immune-driven inflammatory diseases. The molecular mechanisms by which the HO-1 and CO reduce both APC- and T-cell-driven immunity are just beginning to be elucidated. In this article we discuss recent findings related to the immune regulatory capacity of HO-1 and CO at the level of recognition of pathogen-associated molecular patterns and T-cell priming by APCs. Finally, we propose a possible regulatory role for HO-1 and CO over the recently described mitochondria-dependent immunity. These concepts could contribute to the design of new therapeutic tools for inflammation-based diseases.


Assuntos
Apresentação de Antígeno , Heme Oxigenase-1/metabolismo , Doenças do Sistema Imunitário/tratamento farmacológico , Tolerância Imunológica , Inflamação/metabolismo , Linfócitos T/imunologia , Animais , Monóxido de Carbono/metabolismo , Desenho de Fármacos , Humanos , Imunomodulação , Ativação Linfocitária
13.
Eur J Immunol ; 45(12): 3269-88, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26461179

RESUMO

Heme-oxygenase 1 (HO-1) prevents T cell-mediated inflammatory disease by producing carbon monoxide (CO) and impairing DC immunogenicity. However, the cellular mechanisms causing this inhibition are unknown. Here, we show that CO impairs mitochondrial function in DCs by reducing both the mitochondrial membrane potential and ATP production, and resembling the effect of a nonlethal dose of a classical mitochondria uncoupler carbonyl cyanide m-chlorophenyl hydrazone (CCCP). Moreover, both CO and CCCP reduced cargo transport, endosome-to-lysosome fusion, and antigen processing, dampening the production of peptide-MHC complexes on the surface of DCs. As a result, the inhibition of naive CD4(+) T-cell priming was observed. Furthermore, mitochondrial dysfunction in DCs also significantly reduced CD8(+) T cell-dependent type 1 diabetes onset in vivo. These results showed for the first time that CO interferes with T-cell priming by blocking an unknown mitochondria-dependent antigen-processing pathway in mature DC. Interestingly, other immune functions in DCs such as antigen capture, cytokine secretion, costimulation, and cell survival relied on glycolysis, suggesting that oxidative phosphorylation might only play a key role for the maturation of antigen-containing endosomes. In conclusion, CO produced by HO-1 impairs antigen-dependent inflammation by regulating DC immunogenicity by a mitochondria-dependent mechanism.


Assuntos
Apresentação de Antígeno/efeitos dos fármacos , Monóxido de Carbono/farmacologia , Células Dendríticas/imunologia , Endossomos/fisiologia , Mitocôndrias/fisiologia , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Heme Oxigenase-1/fisiologia , Humanos
14.
J Immunol ; 192(3): 1313-9, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24376266

RESUMO

Gap junctions (GJs) mediate intercellular communication between adjacent cells. Previously, we showed that connexin 43 (Cx43), the main GJ protein in the immune system, mediates Ag transfer between human dendritic cells (DCs) and is recruited to the immunological synapse during T cell priming. This crosstalk contributed to T cell activation, intracellular Ca(2+) responses, and cytokine release. However, the role of GJs in NK cell activation by DCs and NK cell-mediated cytotoxicity against tumor cells remains unknown. In this study, we found polarization of Cx43 at the NK/DC and NK/tumor cell-contact sites, accompanied by the formation of functional GJs between NK/DCs and NK/tumor cells, respectively. Cx43-GJ-mediated intercellular communication (GJIC) between human NK and DCs was bidirectional. Blockage of Cx43-GJIC inhibited NK cell activation, though it affected neither the phenotype nor the function of DCs. Cx43 knockdown or inhibition using mimetic peptides greatly reduced CD69 and CD25 expression and IFN-γ release by DC-stimulated NK cells. Moreover, blocking Cx43 strongly inhibited the NK cell-mediated tumor cell lysis associated with inhibition of granzyme B activity and Ca(2+) influx. Our data identify a novel and active role for Cx43-GJIC in human NK cell activation and antitumor effector functions that may be important for the design of new immune therapeutic strategies.


Assuntos
Conexina 43/imunologia , Citotoxicidade Imunológica/imunologia , Células Dendríticas/imunologia , Junções Comunicantes/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Apoptose , Sinalização do Cálcio , Comunicação Celular/imunologia , Linhagem Celular Tumoral , Conexina 43/antagonistas & inibidores , Células Dendríticas/ultraestrutura , Granzimas/fisiologia , Humanos , Vigilância Imunológica , Sinapses Imunológicas/imunologia , Testes de Liberação de Interferon-gama , Células Matadoras Naturais/ultraestrutura
15.
Proc Natl Acad Sci U S A ; 110(22): 9112-7, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23650398

RESUMO

Respiratory syncytial virus (RSV) is the major cause of respiratory illness in infants worldwide. Neurologic alterations, such as seizures and ataxia, have been associated with RSV infection. We demonstrate the presence of RSV proteins and RNA in zones of the brain--such as the hippocampus, ventromedial hypothalamic nucleus, and brainstem--of infected mice. One month after disease resolution, rodents showed behavioral and cognitive impairment in marble burying (MB) and Morris water maze (MWM) tests. Our data indicate that the learning impairment caused by RSV is a result of a deficient induction of long-term potentiation in the hippocampus of infected animals. In addition, immunization with recombinant bacillus Calmette-Guérin (BCG) expressing RSV nucleoprotein prevented behavioral disorders, corroborating the specific effect of RSV infection over the central nervous system. Our findings provide evidence that RSV can spread from the airways to the central nervous system and cause functional alterations to the brain, both of which can be prevented by proper immunization against RSV.


Assuntos
Encéfalo/metabolismo , Deficiências da Aprendizagem/etiologia , RNA Viral/metabolismo , Infecções por Vírus Respiratório Sincicial/complicações , Infecções por Vírus Respiratório Sincicial/imunologia , Proteínas Virais/metabolismo , Animais , Encéfalo/patologia , Deficiências da Aprendizagem/prevenção & controle , Deficiências da Aprendizagem/virologia , Potenciação de Longa Duração/fisiologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium bovis/imunologia , Ratos , Ratos Sprague-Dawley , Infecções por Vírus Respiratório Sincicial/metabolismo , Linfócitos T/imunologia , Vacinas Virais/imunologia
16.
Immunology ; 144(2): 321-32, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25179131

RESUMO

Carbon monoxide (CO) has been recently reported as the main anti-inflammatory mediator of the haem-degrading enzyme haem-oxygenase 1 (HO-1). It has been shown that either HO-1 induction or CO treatment reduces the ability of monocytes to respond to inflammatory stimuli, such as lipopolysaccharide (LPS), due to an inhibition of the signalling pathways leading to nuclear factor-κB, mitogen-activated protein kinases and interferon regulatory factor 3 activation. Hence, it has been suggested that CO impairs the stimulation of the Toll-like receptor 4 (TLR4)/myeloid differentiation factor-2 (MD2) complex located on the surface of immune cells. However, whether CO can negatively modulate the surface expression of the TLR4/MD2 complex in immune cells remains unknown. Here we report that either HO-1 induction or treatment with CO decreases the surface expression of TLR4/MD2 in dendritic cells (DC) and neutrophils. In addition, in a septic shock model of mice intraperitoneally injected with lipopolysaccharide (LPS), prophylactic treatment with CO protected animals from hypothermia, weight loss, mobility loss and death. Further, mice pre-treated with CO and challenged with LPS showed reduced recruitment of DC and neutrophils to peripheral blood, suggesting that this gas causes a systemic tolerance to endotoxin challenge. No differences in the amount of innate cells in lymphoid tissues were observed in CO-treated mice. Our results suggest that CO treatment reduces the expression of the TLR4/MD2 complex on the surface of myeloid cells, which renders them resistant to LPS priming in vitro, as well as in vivo in a model of endotoxic shock.


Assuntos
Monóxido de Carbono/farmacologia , Heme Oxigenase-1/biossíntese , Antígeno 96 de Linfócito/biossíntese , Choque Séptico/prevenção & controle , Receptor 4 Toll-Like/biossíntese , Animais , Antimetabólitos/farmacologia , Movimento Celular/imunologia , Células Dendríticas/imunologia , Ativação Enzimática/imunologia , Hipotermia/tratamento farmacológico , Hipotermia/prevenção & controle , Inflamação/imunologia , Mediadores da Inflamação/farmacologia , Fator Regulador 3 de Interferon/imunologia , Lipopolissacarídeos , Antígeno 96 de Linfócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia , NF-kappa B/imunologia , Neutrófilos/imunologia , Choque Séptico/tratamento farmacológico , Choque Séptico/imunologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/metabolismo
17.
Eur J Immunol ; 43(11): 2832-44, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23852701

RESUMO

Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. We have previously shown that either induction of HO-1 or treatment with exogenous CO inhibits LPS-induced maturation of dendritic cells (DCs) and protects in vivo and in vitro antigen-specific inflammation. Here, we evaluated the capacity of HO-1 and CO to regulate antigen presentation on MHC class I and MHC class II molecules by LPS-treated DCs. We observed that HO-1 and CO treatment significantly inhibited the capacity of DCs to present soluble antigens to T cells. Inhibition was restricted to soluble OVA protein, as no inhibition was observed for antigenic OVA-derived peptides, bead-bound OVA protein, or OVA as an endogenous antigen. Inhibition of soluble antigen presentation was not due to reduced antigen uptake by DCs, as endocytosis remained functional after HO-1 induction and CO treatment. On the contrary, CO significantly reduced the efficiency of fusion between late endosomes and lysosomes and not by phagosomes and lysosomes. These data suggest that HO-1 and CO can inhibit the ability of LPS-treated DCs to present exogenous soluble antigens to naïve T cells by blocking antigen trafficking at the level of late endosome-lysosome fusion.


Assuntos
Apresentação de Antígeno/imunologia , Monóxido de Carbono/metabolismo , Endossomos/metabolismo , Lisossomos/metabolismo , Animais , Apresentação de Antígeno/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Monóxido de Carbono/farmacologia , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Endocitose/imunologia , Endossomos/efeitos dos fármacos , Heme Oxigenase-1/imunologia , Heme Oxigenase-1/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Lipopolissacarídeos/imunologia , Ativação Linfocitária/imunologia , Lisossomos/efeitos dos fármacos , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/imunologia
18.
J Innate Immun ; 16(1): 143-158, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38310854

RESUMO

BACKGROUND: Upon infection, mucosal tissues activate a brisk inflammatory response to clear the pathogen, i.e., resistance to disease. Resistance to disease is orchestrated by tissue-resident macrophages, which undergo profound metabolic reprogramming after sensing the pathogen. These metabolically activated macrophages release many inflammatory factors, which promote their bactericidal function. However, in immunocompetent individuals, pathogens like Pseudomonas aeruginosa, Staphylococcus aureus, and Salmonella evade this type of immunity, generating communities that thrive for the long term. SUMMARY: These organisms develop features that render them less susceptible to eradication, such as biofilms and increased tolerance to antibiotics. Furthermore, after antibiotic therapy withdrawal, "persister" cells rapidly upsurge, triggering inflammatory relapses that worsen host health. How these pathogens persisted in inflamed tissues replete with activated macrophages remains poorly understood. KEY MESSAGES: In this review, we discuss recent findings indicating that the ability of P. aeruginosa, S. aureus, and Salmonella to evolve biofilms and antibiotic tolerance is promoted by the similar metabolic routes that regulate macrophage metabolic reprogramming.


Assuntos
Antibacterianos , Biofilmes , Macrófagos , Biofilmes/efeitos dos fármacos , Humanos , Animais , Macrófagos/imunologia , Macrófagos/microbiologia , Antibacterianos/farmacologia , Infecções Bacterianas/imunologia , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/fisiologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/fisiologia , Farmacorresistência Bacteriana , Evasão da Resposta Imune
19.
Immunology ; 140(1): 123-32, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23691924

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple alterations affecting the normal function of immune cells, such as lymphocytes, dendritic cells (DCs) and monocytes. Although the understanding of autoimmunity has significantly increased, the breakthrough in effective therapies has been modest, making necessary the development of new therapeutic strategies. Here we propose that a new potential target for therapy is haem oxygenase-1 (HO-1), an enzyme that catalyses the degradation of the haem group into biliverdin, carbon monoxide (CO) and Fe(2+) . These products exhibit immunosuppressive and anti-inflammatory effects, which can contribute to improving tolerance during organ transplantation. Because HO-1 is highly expressed by immune cells involved in SLE pathogenesis, such as monocytes and DCs, we evaluated whether induction of HO-1 expression or the administration of CO could ameliorate disease in the FcγRIIb knockout (KO) mouse model for SLE. We found that CO administration decreased the expansion of CD11b(+) cells, prevented the decline of regulatory T cells and reduced anti-histone antibodies observed in untreated FcγRIIb KO mice. Furthermore, CO-treated animals and HO-1 induction showed less kidney damage compared with untreated mice. These data suggest that HO-1 modulation and CO administration can ameliorate autoimmunity and prevent the lupus symptoms shown by FcγRIIb KO mice, highlighting HO-1 as a potential new target for autoimmune therapy.


Assuntos
Monóxido de Carbono/administração & dosagem , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/terapia , Animais , Autoimunidade/efeitos dos fármacos , Antígeno CD11b/metabolismo , Modelos Animais de Doenças , Indução Enzimática/efeitos dos fármacos , Feminino , Heme Oxigenase-1/biossíntese , Rim/efeitos dos fármacos , Rim/enzimologia , Rim/patologia , Lúpus Eritematoso Sistêmico/enzimologia , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de IgG/deficiência , Receptores de IgG/genética , Baço/imunologia , Baço/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
20.
Plant Sci ; 336: 111858, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37673219

RESUMO

The Sirex noctilio's climatic adaption and rapid proliferation have caused Pinus mortality worldwide. The infestation combines the early effect of female S. noctilio gland secretion and the spreading symbiotic fungus Amylostereum areolatum. 'Lipidomics' is the study of all non-water-soluble components of the metabolome. Most of these non-water-soluble compounds correspond to lipids which can provide information about a biological activity, an organelle, an organism, or a disease. Using HPLC-MS/MS based lipidomics, 122 lipids were identified in P. radiata needles during S. noctilio infestation. Phosphatidic acids, N-acylethanolamines, and phosphatidylinositol-ceramides accumulated in infested trees could suggest a high level of phospholipases activities. The phosphatidylcholines were the most down-regulated species during infection, which could also suggest that they may be used as a substrate for up-regulated lipids. The accumulation of very long-chain fatty acids and long-chain fatty acids during the infestation could imply the tree defense response to create a barrier in the drilled zone to avoid larvae development and fungus proliferation. Also, the growth arrest phase of the trees during the prolonged infestation suggests a resistance response, regulated by the accumulation of NAE, which potentially shifts the tree energy to respond to the infestation.


Assuntos
Himenópteros , Pinus , Animais , Metabolismo dos Lipídeos , Lipidômica , Espectrometria de Massas em Tandem , Himenópteros/fisiologia , Fungos , Árvores , Ácidos Graxos , Lipídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA