Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Microb Pathog ; 184: 106357, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37716625

RESUMEN

As a member of the damage-associated molecular patterns, heat shock proteins (HSPs) are widely recognized for their role in initiating innate immune responses. These highly conserved proteins are expressed ubiquitously in both prokaryotes and eukaryotes. In this study, our aim was to investigate how DnaJ, a HSP40 homolog derived from Pseudomonas aeruginosa (P. aeruginosa), influences the regulation of IL-8 expression in macrophages. Treatment with DnaJ served as a stimulus, inducing a more robust expression of IL-8 compared to other HSP homologs, including DnaK, GroEL, and HtpG. This effect was achieved through the activation of the NF-κB signaling pathway. Interestingly, DnaJ treatment also significantly increased the expression of microRNA-146a (miR-146a), which appears to play a role in modulating the expression of innate defense genes. As a consequence, pre-treatment with DnaJ led to a reduction in the extent of IL-8 induction in response to P. aeruginosa treatment. Notably, this reduction was counteracted by transfection of a miR-146a inhibitor, highlighting the involvement of miR-146a in P. aeruginosa-mediated induction of IL-8 expression. Therefore, this study uncovers the role of DnaJ in triggering the expression of miR-146a, which, in turn, modulates the excessive expression of IL-8 induced by P. aeruginosa infection.


Asunto(s)
MicroARNs , MicroARNs/metabolismo , Interleucina-8/genética , FN-kappa B/metabolismo , Transducción de Señal , Macrófagos/metabolismo
2.
Int J Mol Sci ; 24(21)2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37958940

RESUMEN

As members of pathogen-associated molecular patterns, bacterial heat shock proteins (HSPs) are widely recognized for their role in initiating innate immune responses. This study aimed to examine the impact of DnaJ, a homolog of HSP40 derived from Pseudomonas aeruginosa (P. aeruginosa), on the regulation of IL-1ß expression in macrophages. We demonstrated that DnaJ modulates macrophages to secrete IL-1ß by activating NF-κB and MAPK signaling pathways. Specifically, ERK was identified as a positive mediator for IL-1ß expression, while p38 acted as a negative mediator. These results suggest that the reciprocal actions of these two crucial MAPKs play a vital role in controlling IL-1ß expression. Additionally, the reciprocal actions of MAPKs were found to regulate the activation of inflammasome-related molecules, including vimentin, NLRP3, caspase-1, and GSDMD. Furthermore, our investigation explored the involvement of CD91/CD40 in ERK signaling-mediated IL-1ß production from DnaJ-treated macrophages. These findings emphasize the importance of understanding the signaling mechanisms underlying IL-1ß induction and suggest the potential utility of DnaJ as an adjuvant for stimulating inflammasome activation.


Asunto(s)
Inflamasomas , Pseudomonas aeruginosa , Inflamasomas/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal , Macrófagos/metabolismo , FN-kappa B/metabolismo , Interleucina-1beta/metabolismo
3.
Infect Immun ; 90(10): e0032922, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36169312

RESUMEN

Bloodstream infections (BSIs) caused by Pseudomonas aeruginosa are associated with a high mortality rate in the clinic. However, the fitness mechanisms responsible for the evolution of virulence factors that facilitate the dissemination of P. aeruginosa to the bloodstream are poorly understood. In this study, a transcriptomic analysis of the BSI-associated P. aeruginosa clinical isolates showed a high-level expression of cell-surface signaling (CSS) system Hxu. Whole-genome sequencing and comparative genomics of these isolates showed that a mutation in rnfE gene was responsible for the elevated expression of the Hxu-CSS pathway. Most importantly, deletion of the hxuIRA gene cluster in a laboratory strain PAO1 reduced its BSI capability while overexpression of the HxuIRA pathway promoted BSI in a murine sepsis model. We further demonstrated that multiple components in the blood plasma, including heme, hemoglobin, the heme-scavenging proteins haptoglobin, and hemopexin, as well as the iron-delivery protein transferrin, could activate the Hxu system. Together, these studies suggested that the Hxu-CSS system was an important signal transduction pathway contributing to the adaptive pathogenesis of P. aeruginosa in BSI.


Asunto(s)
Infecciones por Pseudomonas , Sepsis , Ratones , Animales , Pseudomonas aeruginosa/metabolismo , Hemopexina/metabolismo , Haptoglobinas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Hemo/metabolismo , Transducción de Señal , Hierro/metabolismo , Hemoglobinas/metabolismo , Transferrinas/metabolismo
4.
Microb Pathog ; 165: 105465, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35247500

RESUMEN

Toll-like receptor 7 (TLR7) signaling plays pivotal roles in innate immunity by sensing viral single-stranded RNA thereby triggering inflammatory signaling cascades and eliciting protective antiviral responses. In this study, we found that TLR7 expression is highly induced in response to Pseudomonas aeruginosa (P. aeruginosa) infection in a dose- and time-dependent manner. P. aeruginosa-derived DnaJ, a homolog of HSP40, was identified as a related inducing agent for TLR7 expression, and expression of DnaJ was stimulated when host cells were infected with P. aeruginosa. Interestingly, DnaJ was not involved in mediating an increase in the expression levels of TLR3 and TLR8, other well-known antiviral receptors. The induction of TLR7 in response to DnaJ was mediated by the activation of the AKT (Thr308 and Ser473)/NF-κB and p38/JNK MAPKs signaling pathways, consequently transmitting related signals for the expression of interferons (IFNs). Of note, these antiviral responses were regulated, at least in part, by TLR4, which senses the presence of DnaJ and then promotes downstream activation of the AKT (Ser473)/NF-κB and JNK signaling cascades. Taken together, these results suggest that P. aeruginosa-derived DnaJ is sufficient to promote an increase in TLR7 expression in the TLR4-engaged AKT/NF-κB and JNK signaling pathways, thereby promoting an increased antiviral response through the elevated expression of IFNs.


Asunto(s)
FN-kappa B , Receptor Toll-Like 7 , Antivirales , Interferones/metabolismo , Sistema de Señalización de MAP Quinasas , Macrófagos/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pseudomonas aeruginosa/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
5.
Nutr Cancer ; 73(10): 2078-2088, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32964733

RESUMEN

Cancer is caused by uncontrolled cell division and is a leading cause of mortality worldwide. Oenothera odorata (O. odorata) extract is used in herbal medicine to inhibit inflammation, but its potential anti-tumor properties have not been fully evaluated. Here, we demonstrated that O. odorata extract inhibits the proliferation of lung adenocarcinoma and melanoma cell lines In Vitro, and also inhibits the growth of melanoma cells In Vivo. After partitioning the extract with n-hexane, chloroform, ethyl acetate, and n-butanol, it was found that the butanol-soluble (OOB) and water-soluble (OOW) fractions of O. odorata extract are effective at inhibiting tumor cell growth In Vivo although OOW is more effective than OOB. Interestingly, these fractions did not inhibit the growth of non-cancerous cells. The anti-proliferative effects of the OOW fraction were found to be mediated by inhibition of glycolysis and cellular respiration. UPLC of both fractions showed two major common peaks, which were predicted to be hydrolyzable tannin-related compounds. Taken together, these data suggest that O. odorata extract has anti-tumor properties, and the molecular mechanism involves metabolic alterations and inhibition of cell proliferation. O. odorata extract therefore holds promise as a novel natural product for the treatment of cancer.


Asunto(s)
Neoplasias , Oenothera , Plantas Medicinales , Respiración de la Célula , Glucólisis , Extractos Vegetales/farmacología
6.
Int J Mol Sci ; 21(23)2020 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-33276561

RESUMEN

IL-27, a heterodimeric cytokine composed of the p28 subunit and Epstein-Barr virus-induced gene 3 (EBI3), acts as a potent immunosuppressant and thus limits pathogenic inflammatory responses. IL-27 is upregulated upon Pseudomonas aeruginosa infection in septic mice, increasing susceptibility to the infection and decreasing clearance of the pathogen. However, it remains unclear which P. aeruginosa-derived molecules promote production of IL-27. In this study, we explored the mechanism by which P. aeruginosa DnaK, a heat shock protein 70-like protein, induces EBI3 expression, thereby promoting production of IL-27. Upregulation of EBI3 expression did not lead to an increase in IL-35, which consists of the p35 subunit and EBI3. The IL-27 production in response to DnaK was biologically active, as reflected by stimulation of IL-10 production. DnaK-mediated expression of EBI3 was driven by two distinct signaling pathways, NF-κB and Akt. However, NF-κB is linked to TLR4-associated signaling pathways, whereas Akt is not. Taken together, our results reveal that P. aeruginosa DnaK potently upregulates EBI3 expression, which in turn drives production of the prominent anti-inflammatory cytokine IL-27, as a consequence of TLR4-dependent activation of NF-κB and TLR4-independent activation of the Akt signaling pathway.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Interleucina-27/biosíntesis , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/metabolismo , Proteínas Bacterianas/genética , Regulación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Interacciones Huésped-Patógeno , Interleucinas/genética , Interleucinas/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pseudomonas aeruginosa/genética , Transducción de Señal , Receptor Toll-Like 4/metabolismo
7.
Infect Immun ; 85(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28031262

RESUMEN

As one of the first lines of host defense, monocytes play important roles in clearing infected microbes. The defensive response is triggered by recognition of diverse microbial moieties, including released factors, which modulate host immune responses to establish a harsh environment for clinically important bacterial pathogens. In this study, we found that the expression of PTX3, a soluble form of pattern recognition receptor, was induced by infection with live Pseudomonas aeruginosa or treatment of cells with its supernatant. P. aeruginosa GroEL, a homolog of heat shock protein 60, was identified as one of the factors responsible for inducing the expression of PTX3 in host cells. GroEL induced PTX3 expression by activating the Toll-like receptor 4 (TLR4)-dependent pathway via nuclear factor-kappa B (NF-κB), while simultaneously inhibiting expression of microRNA-9, which targets the PTX3 transcript. Finally, by acting as an opsonin, GroEL-induced PTX3 promoted the association and phagocytosis of Staphylococcus aureus into macrophages. These data suggest that the host defensive environment is supported by the production of PTX3 in response to GroEL, which thus has therapeutic potential for clearance of bacterial infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteína C-Reactiva/biosíntesis , MicroARNs/genética , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiología , Componente Amiloide P Sérico/biosíntesis , Transducción de Señal , Proteína C-Reactiva/genética , Línea Celular , Células Cultivadas , Regulación de la Expresión Génica , Humanos , FN-kappa B/metabolismo , Fagocitosis , Infecciones por Pseudomonas/microbiología , Componente Amiloide P Sérico/genética , Receptor Toll-Like 4/metabolismo
8.
J Infect Dis ; 214(6): 962-9, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27377745

RESUMEN

The type III secretion system (T3SS) in Pseudomonas aeruginosa has been linked to severe disease and poor clinical outcomes in animal and human studies. We aimed to investigate whether the ExoS and ExoT effector proteins of P. aeruginosa affect the expression of matrix metalloproteinase 12 (MMP-12) and MMP-13 via nuclear factor κB (NF-κB) signaling pathways. To understand the T3SS, we used ΔExoS, ΔExoT, and ExsA::Ω mutants, as well as P. aeruginosa strain K (PAK)-stimulated NCI-H292 cells. We investigated the effects of ΔExoS, ΔExoT, and ExsA::Ω on the development of pneumonia in mouse models. We examined the effects of ΔExoS, ΔExoT, and ExsA::Ω on MMP-12 and MMP-13 production in NCI-H292 cells. ΔExoS and ΔExoT markedly decreased the neutrophil count in bronchoalveolar lavage fluid, with a reduction in proinflammatory mediators, MMP-12, and MMP-13. ΔExoS and ΔExoT reduced NF-κB phosphorylation, together with MMP-12 and MMP-13 expression in PAK-infected mouse models and NCI-H292 cells. To conclude, P. aeruginosa infection induced the expression of MMPs, and P. aeruginosa T3SS appeared to be a key player in MMP-12 and MMP-13 expression, which is further controlled by NF-κB signaling. These findings might be useful in devising a novel therapeutic approach to chronic pulmonary infections that involves decreasing the ExoS and ExoT levels.


Asunto(s)
Células Epiteliales/microbiología , Metaloproteinasa 12 de la Matriz/biosíntesis , Metaloproteinasa 13 de la Matriz/biosíntesis , FN-kappa B/metabolismo , Neumonía Bacteriana/patología , Pseudomonas aeruginosa/patogenicidad , Sistemas de Secreción Tipo III/metabolismo , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Técnicas de Inactivación de Genes , Interacciones Huésped-Patógeno , Humanos , Metaloproteinasa 12 de la Matriz/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Neumonía Bacteriana/microbiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transactivadores/genética , Transactivadores/metabolismo
9.
Infect Immun ; 83(12): 4791-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26438797

RESUMEN

Pseudomonas aeruginosa infections persist in patients with cystic fibrosis (CF) and drive lung disease progression. P. aeruginosa potently activates the innate immune system mostly through the recognition of pathogen-associated molecular patterns, such as flagellin. Matrix metalloproteinases 12 and 13 (MMP-12 and MMP-13, respectively) exacerbate chronic lung infection and inflammation by promoting uncontrolled tissue rearrangements and fibrosis, yet the underlying molecular mechanisms by which this occurs remain largely unknown. In this study, we used quantitative bacteriology, histological examination, and proinflammatory cytokine levels to evaluate the effects of MMP-12 and MMP-13 on P. aeruginosa strain K-induced infection and pneumonia in H292 epithelial cells and mice, respectively. Under inflammatory stimulation, mRNA and protein expression levels of proinflammatory mediators were higher in strain K-infected mice and cells than in uninfected counterparts, in which MMP-12 and MMP-13 expression reached levels similar to those observed in epithelial cells. Moreover, we also found that the NF-κB pathway might be involved in the induction of cytokines in response to strain K infection. Taken together, these data suggest that MMP-12 and MMP-13 alter strain K infection in mice and play a role in inflammatory regulation by modulating cytokine levels.


Asunto(s)
Metaloproteinasa 12 de la Matriz/inmunología , Metaloproteinasa 13 de la Matriz/inmunología , Neumonía/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/patogenicidad , Animales , Línea Celular Tumoral , Fibrosis Quística/genética , Fibrosis Quística/inmunología , Fibrosis Quística/microbiología , Fibrosis Quística/patología , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Células Epiteliales/patología , Femenino , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Metaloproteinasa 12 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/inmunología , Neumonía/genética , Neumonía/microbiología , Neumonía/patología , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/inmunología , Transducción de Señal
10.
Infect Immun ; 82(8): 3252-60, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24866792

RESUMEN

Inflammatory responses are a first line of host defense against a range of invading pathogens, consisting of the release of proinflammatory cytokines followed by attraction of polymorphonuclear neutrophils (PMNs) to the site of inflammation. Among the many virulence factors that contribute to the pathogenesis of infections, nucleoside diphosphate kinase (Ndk) mediates bacterially induced toxicity against eukaryotic cells. However, no study has examined how Ndk affects inflammatory responses. The present study examined the mechanisms by which Pseudomonas aeruginosa activates inflammatory responses upon infection of cells. The results showed that bacterial Ndk, with the aid of an additional bacterial factor, flagellin, induced expression of the proinflammatory cytokines interleukin-1α (IL-1α) and IL-1ß. Cytokine induction appeared to be dependent on the kinase activity of Ndk and was mediated via the NF-κB signaling pathway. Notably, Ndk activated the Akt signaling pathway, which acts upstream of NF-κB, as well as caspase-1, which is a key component of inflammasome. Thus, this study demonstrated that P. aeruginosa, through the combined effects of Ndk and flagellin, upregulates the expression of proinflammatory cytokines via the Akt/NF-κB signaling pathways.


Asunto(s)
Flagelina/inmunología , Interleucina-1/biosíntesis , Nucleósido-Difosfato Quinasa/inmunología , Pseudomonas aeruginosa/inmunología , Transducción de Señal , Línea Celular , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
11.
Microbiology (Reading) ; 160(Pt 7): 1417-1426, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24699069

RESUMEN

Pseudomonas aeruginosa is a Gram-negative opportunistic human pathogen possessing a type III secretion system (T3SS) which injects toxic effector proteins into mammalian host cells. In previous studies, P. aeruginosa strains lacking all of the known type III effectors were shown to cause cytotoxicity upon prolonged infection time. In this study, we report the identification of a new cytotoxin, nucleoside diphosphate kinase (NDK), which is injected into eukaryotic cells in a T3SS-dependent manner. Injection of NDK is inhibited by the presence of previously known effectors of the T3SS, with an effectorless strain injecting the highest amount, suggesting active competition with the known T3SS effectors. NDK is shown to cause a cytotoxic response when expressed in eukaryotic cells, and P. aeruginosa strains harbouring NDK also show a greater toxicity than strains lacking it. Interestingly, the cytotoxic effect of intracellular NDK is independent of its kinase activity. In previous studies, NDK was shown to be secreted into culture supernatants via a type I secretion system and cause cytotoxicity in a kinase-dependent manner. Therefore, the current study highlights an alternative route of NDK secretion as well as two different cytotoxic mechanisms of NDK, depending on the extra- or intra-cellular location of the protein.


Asunto(s)
Sistemas de Secreción Bacterianos , Toxinas Bacterianas/metabolismo , Nucleósido-Difosfato Quinasa/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Transporte Biológico , Supervivencia Celular , Femenino , Genes Reporteros , Células HeLa , Humanos , Nucleósido-Difosfato Quinasa/genética , Plásmidos/genética , Pseudomonas aeruginosa/genética , Proteínas Recombinantes de Fusión , Eliminación de Secuencia
12.
Microb Pathog ; 66: 24-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24361345

RESUMEN

Infection with Pseudomonas aeruginosa results in a massive accumulation of neutrophils in response to prolonged and sustained expression of inflammatory mediators. The major chemokine associated with this excessive neutrophil recruitment is IL-8, the accumulation of which is a hallmark of cornea and cystic fibrosis airway inflammation. To date, several P. aeruginosa-associated and extracellular factors required for the stimulation of IL-8 expression have been identified. Here, we report a novel effector molecule, nucleoside diphosphate kinase (Ndk), which increases the expression of IL-8 by translocating into host cells. The induction appears to be dependent on both the kinase activity of Ndk and an additional bacterial factor, flagellin, via an NF-κB signaling pathway. This study demonstrates the role of a novel effector, Ndk, which is capable of inducing prominent inflammatory chemokine IL-8 expression with the aid of flagellin during P. aeruginosa infection.


Asunto(s)
Células Epiteliales/microbiología , Flagelos/metabolismo , Interleucina-8/metabolismo , Nucleósido-Difosfato Quinasa/metabolismo , Pseudomonas aeruginosa/genética , Células Cultivadas , Células Epiteliales/metabolismo , Flagelina/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Interleucina-8/genética , FN-kappa B/metabolismo , Infiltración Neutrófila , Neutrófilos/metabolismo , Nucleósido-Difosfato Quinasa/genética , Pseudomonas aeruginosa/metabolismo , Transducción de Señal , Regulación hacia Arriba
13.
Analyst ; 139(15): 3781-9, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-24909145

RESUMEN

A label-free immunosensing method based on the conformational change of calcium-binding protein (CBP) depending on analyte concentration was explored for semi-continuous analysis of free Ca(2+). Glucose-galactose-binding protein as a CBP and produced as a recombinant protein by Escherichia coli was used as the immunogen to produce monoclonal antibodies by hybridoma technology. We finally screened the 3-6F cell clone, which produced the desired antibody specific to a particular structural conformation of the protein that occurred only upon CBP-calcium complex formation. To construct an immunosensor, the antibody was immobilized via a secondary antibody on an Octet Red optical fiber-based label-free sensor. Calcium analysis was conducted on the sensor in combination with CBP previously added to the aqueous sample, which distinguished the sensor signal according to the analyte concentration. The immunosensor produced a signal in real time with a response time of approximately 15 min and could be reused for analyses of different samples in a semi-continuous manner. The minimum detection limit of the analyte under optimal conditions was 0.09 mM and the upper limit was about 5 mM (log-logit transformed standard curve linearity: R(2) > 98%). In sample tests with milk, the analytical performance of the sensor was highly correlated (R(2) > 99%) with that of the reference system based on the KMnO4 titration method (ISO 12081). Although the sensor showed cross-reactivity at high concentrations (>1 mM) of cations including zinc, iron, manganese, and copper, these ionic components were not traceable (<0.01 mM) in milk.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Calcio/análisis , Tecnología de Fibra Óptica/métodos , Inmunoensayo/métodos , Leche/química , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas de Unión Periplasmáticas/metabolismo , Animales , Anticuerpos Inmovilizados/inmunología , Calcio/metabolismo , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/inmunología , Ratones , Proteínas de Transporte de Monosacáridos/química , Proteínas de Transporte de Monosacáridos/inmunología , Fibras Ópticas , Proteínas de Unión Periplasmáticas/química , Proteínas de Unión Periplasmáticas/inmunología , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo
14.
Microbiol Spectr ; 11(3): e0042623, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37039709

RESUMEN

Polymyxins are currently the last-resort antibiotics for the treatment of multidrug-resistant Gram-negative bacterial infections. To expand the understanding of the intrinsic resistance mechanism against polymyxins, a laboratory strain of Pseudomonas aeruginosa PAO1 was subjected to serial passage in the presence of sublethal doses of polymyxin B over a period of 30 days. By whole-genome sequencing of successively isolated polymyxin B-resistant isolates, we identified a frameshift mutation (L183fs) in the mvfR gene that further increased polymyxin resistance in the pmrB mutant background. A ΔmvfR mutation alone showed higher tolerance to polymyxin B due to altered lipopolysaccharide (LPS) on the surface of bacterial cells, which decreases its outer membrane permeability. In the ΔmvfR mutant, polymyxin B treatment caused the upregulation of rfaD, the gene involved in LPS core oligosaccharide synthesis, which is responsible for polymyxin tolerance. To the best of our knowledge, this is the first report of mvfR mutation conferring polymyxin resistance in P. aeruginosa via increased integrity of bacterial outer membrane. IMPORTANCE Antibiotic resistance imposes a considerable challenge for the treatment of P. aeruginosa infections. Polymyxins are the last-resort antibiotics for the treatment of multidrug-resistant P. aeruginosa infections. Understanding the development and mechanisms of bacterial resistance to polymyxins may provide clues for the development of new or improved therapeutic strategies effective against P. aeruginosa. In this study, using an in vitro evolution assay in combination with whole-genome sequencing, we demonstrated that MvfR controls tolerance to polymyxin B by regulating the rfaD gene in P. aeruginosa. Our results reveal a novel mechanism employed by P. aeruginosa in the defense against polymyxin antibiotics.


Asunto(s)
Polimixina B , Pseudomonas aeruginosa , Polimixina B/farmacología , Lipopolisacáridos , Antibacterianos/farmacología , Polimixinas/farmacología , Pruebas de Sensibilidad Microbiana
15.
Front Cell Infect Microbiol ; 13: 1267748, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38029243

RESUMEN

Pseudomonas aeruginosa is an opportunistic human pathogen capable of causing a variety of acute and chronic infections. Its type III secretion system (T3SS) plays a critical role in pathogenesis during acute infection. ExsA is a master regulator that activates the expression of all T3SS genes. Transcription of exsA is driven by two distinct promoters, its own promoter PexsA and its operon promoter PexsC. Here, in combination with a DNA pull-down assay and mass spectrometric analysis, we found that a histone-like nucleoid-structuring (H-NS) family protein MvaT can bind to the PexsC promoter. Using EMSA and reporter assays, we further found that MvaT directly binds to the PexsC promoter to repress the expression of T3SS genes. The repression of MvaT on PexsC is independent of ExsA, with MvaT binding to the -429 to -380 bp region relative to the transcription start site of the exsC gene. The presented work further reveals the complex regulatory network of the T3SS in P. aeruginosa.


Asunto(s)
Transactivadores , Sistemas de Secreción Tipo III , Humanos , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo , Transactivadores/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Proteínas Bacterianas/metabolismo , Regiones Promotoras Genéticas , Regulación Bacteriana de la Expresión Génica
16.
Front Immunol ; 14: 1129705, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36895557

RESUMEN

COVID-19 pandemic continues to spread throughout the world with an urgent demand for a safe and protective vaccine to effectuate herd protection and control the spread of SARS-CoV-2. Here, we report the development of a bacterial vector COVID-19 vaccine (aPA-RBD) that carries the gene for the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein. Live-attenuated strains of Pseudomonas aeruginosa (aPA) were constructed which express the recombinant RBD and effectively deliver RBD protein into various antigen presenting cells through bacterial type 3 secretion system (T3SS) in vitro. In mice, two-dose of intranasal aPA-RBD vaccinations elicited the development of RBD-specific serum IgG and IgM. Importantly, the sera from the immunized mice were able to neutralize host cell infections by SARS-CoV-2 pseudovirus as well as the authentic virus variants potently. T-cell responses of immunized mice were assessed by enzyme-linked immunospot (ELISPOT) and intracellular cytokine staining (ICS) assays. aPA-RBD vaccinations can elicit RBD-specific CD4+and CD8+T cell responses. T3SS-based RBD intracellular delivery heightens the efficiency of antigen presentation and enables the aPA-RBD vaccine to elicit CD8+T cell response. Thus, aPA vector has the potential as an inexpensive, readily manufactured, and respiratory tract vaccination route vaccine platform for other pathogens.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Animales , Humanos , Ratones , Sistemas de Secreción Tipo III , COVID-19/prevención & control , Pandemias , SARS-CoV-2
17.
Anal Chem ; 84(22): 9713-20, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23101931

RESUMEN

In this study, a novel mammalian cell receptor-based immuno-analytical method was developed for the detection of food-poisoning microorganisms by employing toll-like receptors (TLRs) as sensing elements. Upon infection with bacterium, the host cells respond by expressing TLRs, particularly TLR1, TLR2, and TLR4, on the outer membrane surfaces. To demonstrate the potential of using this method for detection of foodborne bacteria, we initially selected two model sensing systems, expression of TLR1 on a cell line, A549, for Escherichia coli and TLR2 on a cell line, RAW264.7, for Shigella sonnei (S. sonnei). Each TLR was detected using antibodies specific to the respective marker. We also found that the addition of immunoassay for the pathogen captured by the TLRs on the mammalian cells significantly enhanced the detection capability. A dual-analytical system for S. sonnei was constructed and successfully detected an extremely low number (about 3.2 CFU per well) of the pathogenic bacterium 5.1 h after infection. This detection time was 2.5 h earlier than the time required for detection using the conventional immunoassay. To endow the specificity of detection, the target bacterium was immuno-magnetically concentrated by a factor of 50 prior to infection. This further shortened the response to approximately 3.4 h, which was less than half of the time needed when the conventional method was used. Such enhanced performance could basically result from synergistic effects of bacterial dose increase and subsequent autocrine signaling on TLRs' up-regulation upon infection with live bacterium. This TLR-based immuno-sensing approach may also be expanded to monitor infection of the body, provided scanning of the signal is feasible.


Asunto(s)
Técnicas Biosensibles/métodos , Escherichia coli/aislamiento & purificación , Inmunoensayo/métodos , Shigella sonnei/aislamiento & purificación , Receptores Toll-Like/metabolismo , Animales , Línea Celular , Escherichia coli/inmunología , Escherichia coli/fisiología , Regulación de la Expresión Génica/inmunología , Humanos , Inmunidad Innata , Imanes/química , Shigella sonnei/inmunología , Shigella sonnei/fisiología
18.
J Microbiol ; 60(6): 649-656, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35489019

RESUMEN

Inflammatory responses involve the action of inflammatory mediators that are necessary for the clearance of invading bacterial pathogens. However, excessive production of inflammatory mediators can damage tissues, thereby impairing bacterial clearance. Here, we examined the effects of Weigela florida on the expression of inflammatory cytokines induced by Pseudomonas aeruginosa or Staphylococcus aureus infection in macrophages. The results showed that pre-treatment with W. florida markedly downregulated the bacterial infection-mediated expression of cytokines. Additionally, post-treatment also triggered anti-inflammatory effects in cells infected with S. aureus to a greater extent than in those infected with P. aeruginosa. Bacterial infection activated inflammation-associated AKT (Thr308 and Ser473)/NF-κB and MAPK (p38, JNK, and ERK) signaling pathways, whereas W. florida treatment typically inhibited the phosphorylation of AKT/NF-κB and p38/JNK, supporting the anti-inflammatory effects of W. florida. The present results suggest that W. florida decreases the infection-mediated expression of inflammatory mediators by inhibiting the AKT/NF-κB and MAPK signaling pathways, implying that it may have potential use as an inhibitory agent of excessive inflammatory responses.


Asunto(s)
Mediadores de Inflamación , Infecciones Estafilocócicas , Antiinflamatorios/farmacología , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/farmacología , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pseudomonas aeruginosa/metabolismo , Staphylococcus aureus
19.
mBio ; 13(3): e0054722, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35467416

RESUMEN

Pseudomonas aeruginosa is a ubiquitous pathogenic bacterium that can adapt to a variety environments. The ability to effectively sense and respond to host local nutrients is critical for the infection of P. aeruginosa. However, the mechanisms employed by the bacterium to respond to nutrients remain to be explored. CspA family proteins are RNA binding proteins that are involved in gene regulation. We previously demonstrated that the P. aeruginosa CspA family protein CspC regulates the type III secretion system in response to temperature shift. In this study, we found that CspC regulates the quorum-sensing (QS) systems by repressing the translation of a QS negative regulatory gene, rsaL. Through RNA immunoprecipitation coupled with real-time quantitative reverse transcription-PCR (RIP-qRT-PCR) and electrophoretic mobility shift assays (EMSAs), we found that CspC binds to the 5' untranslated region of the rsaL mRNA. Unlike glucose, itaconate (a metabolite generated by macrophages during infection) reduces the acetylation of CspC, which increases the affinity between CspC and the rsaL mRNA, leading to upregulation of the QS systems. Our results revealed a novel regulatory mechanism of the QS systems in response to a host-generated metabolite. IMPORTANCE Bacterial infectious diseases impose a severe threat to human health. The ability to orchestrate virulence determinant in response to the host environment is critical for the pathogenesis of bacterial pathogens. Pseudomonas aeruginosa is a leading pathogen that causes various infections in humans. In P. aeruginosa, the quorum-sensing (QS) systems play an important role in regulating the production of virulence factors. In this study, we find that a small RNA binding protein, CspC, regulates the QS systems by repressing the expression of a QS negative regulator. We further demonstrate that CspC is acetylated in response to a host-derived metabolite, itaconate, which alters the function of CspC in regulating the QS system. The importance of this work is in elucidation of a novel regulatory pathway that regulates virulence determinants in P. aeruginosa in response to a host signal.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Pseudomonas aeruginosa , Acetilación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Humanos , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum/genética , ARN Mensajero/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
20.
Microbiol Spectr ; 10(1): e0162021, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35044199

RESUMEN

The opportunistic pathogen Pseudomonas aeruginosa often adapts to its host environment and causes recurrent nosocomial infections. The extracytoplasmic function (ECF) sigma factor enables bacteria to alter their gene expression in response to host environmental stimuli. Here, we report an ECF sigma factor, HxuI, which is rapidly induced once P. aeruginosa encounters the host. Host stresses such as iron limitation, oxidative stress, low oxygen, and nitric oxide induce the expression of hxuI. By combining RNA-seq and promoter-lacZ reporter fusion analysis, we reveal that HxuI can activate the expression of diverse metabolic and virulence pathways which are critical to P. aeruginosa infections, including iron acquisition, denitrification, pyocyanin synthesis, and bacteriocin production. Most importantly, overexpression of the hxuI in the laboratory strain PAO1 promotes its colonization in both murine lung and subcutaneous infections. Together, our findings show that HxuI, a key player in host stress-response, controls the in vivo adaptability and virulence of P. aeruginosa during infection. IMPORTANCE P. aeruginosa has a strong ability to adapt to diverse environments, making it capable of causing recurrent and multisite infections in clinics. Understanding host adaptive mechanisms plays an important guiding role in the development of new anti-infective agents. Here, we demonstrate that an ECFσ factor of P. aeruginosa response to the host-inflicted stresses, which promotes the bacterial in vivo fitness and pathogenicity. Furthermore, our findings may help explain the emergence of highly transmissible strains of P. aeruginosa and the acute exacerbations during chronic infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/metabolismo , Factor sigma/metabolismo , Animales , Proteínas Bacterianas/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Pseudomonas aeruginosa/genética , Regulón , Factor sigma/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA