Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Exp Eye Res ; 179: 1-7, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30343040

RESUMO

Research with animal models of Pseudomonas aeruginosa keratitis has shown that use of a topical corticosteroid alone against an established infection can significantly increase the number of colonizing bacteria or worsen clinical disease. Moreover, retrospective analysis has suggested that corticosteroid use in humans is associated with an increased risk of keratitis in eyes with pre-existing disease. Thus, while corticosteroids are often used to reduce ocular inflammation in the absence of infection, the risk of opportunistic infection remains a concern. However, the effect of corticosteroids on the intrinsic barrier function of uninfected corneas is unknown. Here, we tested if short-term topical corticosteroid treatment of an uninfected murine cornea would increase susceptibility to P. aeruginosa colonization or infection after epithelial injury. Topical prednisolone acetate (1%) was administered to one eye of C57BL/6 mice three times a day for 3 days; control eyes were treated with sterile PBS. Prior to inoculation with a cytotoxic P. aeruginosa corneal isolate strain 6206, corneas were subject to superficial-injury by tissue paper blotting, or scratch-injured followed by 12 h of healing. Previously we have shown that blotting renders mouse corneas susceptible to P. aeruginosa adhesion, but not infection, while 12 h healing reduces susceptibility to infection after scratching. Corneas were evaluated at 48 h for bacterial colonization and microbial keratitis (MK). To monitor impact on wound healing, corneal integrity was examined by fluorescein staining immediately after scarification and after 12 h healing. For both the tissue paper blotting and scratch-injury models, there was no significant difference in P. aeruginosa colonization at 48 h between corticosteroid-pretreated eyes and controls. With the blotting model, one case of MK was observed in a control (PBS-pretreated) cornea; none in corticosteroid-pretreated corneas. With the 12 h healing model, MK occurred in 6 of 17 corticosteroid-pretreated eyes versus 2 of 17 controls, a difference not statistically significant. Corticosteroid-pretreated eyes showed greater fluorescein staining 12 h after scarification injury, but this did not coincide with increased colonization or MK. Together, these data show that short-term topical corticosteroid therapy on an uninfected murine cornea does not necessarily enhance its susceptibility to P. aeruginosa colonization or infection after injury, even when it induces fluorescein staining.


Assuntos
Lesões da Córnea/microbiologia , Infecções Oculares Bacterianas/microbiologia , Glucocorticoides/uso terapêutico , Prednisolona/análogos & derivados , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/isolamento & purificação , Administração Oftálmica , Animais , Córnea/efeitos dos fármacos , Lesões da Córnea/diagnóstico , Úlcera da Córnea/diagnóstico , Úlcera da Córnea/microbiologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Epitélio Corneano/lesões , Infecções Oculares Bacterianas/diagnóstico , Feminino , Fluoresceína/metabolismo , Corantes Fluorescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Prednisolona/uso terapêutico , Pré-Medicação , Infecções por Pseudomonas/diagnóstico , Estudos Retrospectivos , Cicatrização
2.
Sci Transl Med ; 15(686): eade2909, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36888696

RESUMO

Controlling infection-driven inflammation is a major clinical dilemma because of limited therapeutic options and possible adverse effects on microbial clearance. Compounding this difficulty is the continued emergence of drug-resistant bacteria, where experimental strategies aiming to augment inflammatory responses for enhanced microbial killing are not applicable treatment options for infections of vulnerable organs. As with corneal infections, severe or prolonged inflammation jeopardizes corneal transparency, leading to devastating vision loss. We hypothesized that keratin 6a-derived antimicrobial peptides (KAMPs) may be a two-pronged remedy capable of tackling bacterial infection and inflammation at once. We used murine peritoneal neutrophils and macrophages, together with an in vivo model of sterile corneal inflammation, to find that nontoxic and prohealing KAMPs with natural 10- and 18-amino acid sequences suppressed lipoteichoic acid (LTA)- and lipopolysaccharide (LPS)-induced NFκB and IRF3 activation, proinflammatory cytokine production, and phagocyte recruitment independently of their bactericidal function. Mechanistically, KAMPs not only competed with bacterial ligands for cell surface Toll-like receptor (TLR) and co-receptors (MD2, CD14, and TLR2) but also reduced cell surface availability of TLR2 and TLR4 through promotion of receptor endocytosis. Topical KAMP treatment effectively alleviated experimental bacterial keratitis, as evidenced by substantial reductions of corneal opacification, inflammatory cell infiltration, and bacterial burden. These findings reveal the TLR-targeting activities of KAMPs and demonstrate their therapeutic potential as a multifunctional drug for managing infectious inflammatory disease.


Assuntos
Queratinas , Receptor 2 Toll-Like , Camundongos , Animais , Receptor 2 Toll-Like/metabolismo , Queratinas/metabolismo , Inflamação/tratamento farmacológico , Peptídeos/metabolismo , Proteínas de Transporte , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Receptores de Lipopolissacarídeos/metabolismo
3.
Infect Immun ; 79(2): 595-605, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21115716

RESUMO

Studies have shown that epithelium-expressed antimicrobial peptides (AMPs), e.g., ß-defensins, play a role in clearing bacteria from mouse corneas already infected with Pseudomonas aeruginosa. Less is known about the role of AMPs in allowing the cornea to resist infection when healthy. We previously reported that contact lens exposure, a major cause of P. aeruginosa keratitis, can inhibit the upregulation of human ß-defensin 2 (hBD-2) by corneal epithelial cells in response to P. aeruginosa antigens in vitro. Here, we studied the role of AMPs in maintaining the corneal epithelial barrier to P. aeruginosa penetration using both in vitro (human) and in vivo (mouse) experiments. Results showed that preexposing human corneal epithelial multilayers to bacterial antigens in a culture supernatant (known to upregulate AMP expression) reduced epithelial susceptibility to P. aeruginosa traversal up to 6-fold (P < 0.001). Accordingly, small interfering RNA (siRNA) knockdown of any one of four AMPs expressed by human epithelia promoted P. aeruginosa traversal by more than 3-fold (P < 0.001). The combination knockdown of AMPs further enhanced susceptibility to bacterial traversal by ∼8-fold (P < 0.001). In vivo experiments showed that the loss of murine ß-defensin 3 (mBD-3), a murine ortholog of hBD-2, enhanced corneal susceptibility to P. aeruginosa. The uninjured ocular surface of mBD-3(-/-) mice showed a reduced capacity to clear P. aeruginosa, and their corneal epithelia were more susceptible to bacterial colonization, even when inoculated ex vivo to exclude tear fluid effects. Together, these in vitro and in vivo data show functional roles for AMPs in normal corneal epithelial cell barrier function against P. aeruginosa.


Assuntos
Defensinas/metabolismo , Epitélio Corneano/metabolismo , Epitélio Corneano/microbiologia , Pseudomonas aeruginosa/fisiologia , Animais , Linhagem Celular , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , beta-Defensinas/genética , beta-Defensinas/metabolismo
4.
Microb Pathog ; 51(5): 305-12, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21843628

RESUMO

We previously showed that ADP-ribosylation (ADP-r) activity of ExoS, a type III secreted toxin of Pseudomonas aeruginosa, enables bacterial replication in corneal and respiratory epithelial cells and correlates with bacterial trafficking to plasma membrane blebs (bleb-niche formation). Here, we explored another type III secreted toxin, ExoY, for its impact on intracellular trafficking and survival, and for virulence in vivo using a murine corneal infection model. Chromosomal or plasmid-mediated expression of exoY in invasive P. aeruginosa (strain PAO1) enabled bacteria to form and traffic to epithelial membrane blebs in the absence of other known effectors. In contrast, plasmid expression of any of four adenylate cyclase mutant forms of exoY did not enable bleb-niche formation, and bacteria localized to perinuclear vacuoles as for effector-null mutant controls. None of the plasmid-complemented bacteria used in this study showed ADP-r activity in the absence of ExoS and ExoT. In contrast to ADP-r activity of ExoS, bleb-niche formation induced by ExoY's adenylate cyclase activity was not accompanied by enhanced intracellular replication. In vivo results showed that ExoY-adenylate cyclase activity promoted P. aeruginosa corneal virulence in susceptible mice. Together the data show that adenylate cyclase activity of P. aeruginosa ExoY, similarly to the ADP-r activity of ExoS, can mediate bleb-niche formation in epithelial cells. While this activity did not promote intracellular replication in vitro, ExoY conferred increased virulence in vivo in susceptible mice. Mechanisms for bleb-niche formation and relationships to intracellular replication and virulence in vivo require further investigation for both ExoS and ExoY.


Assuntos
Adenilil Ciclases/metabolismo , Proteínas de Bactérias/metabolismo , Células Epiteliais/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/patogenicidade , Adenilil Ciclases/química , Adenilil Ciclases/genética , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Linhagem Celular , Membrana Celular/microbiologia , Células Epiteliais/citologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/genética , Virulência
5.
J Control Release ; 333: 283-297, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33798665

RESUMO

Tacrolimus has shown efficacy in eye inflammatory diseases. However, due to the drug lability, its formulation into a stable ophthalmic product remains a challenge. Tacrolimus-loaded nanocapsules (NCs) were designed for ocular instillation. Further, the stability and effects of the formulation were analyzed under different experimental conditions. Physicochemical characterization of the NCs revealed suitable homogeneous size and high encapsulation efficiency. Moreover, the lyophilized formulation was stable at ICH long term and accelerated storage conditions, for at least 18 and 3 months, respectively. The tacrolimus NCs did not elicit any eye irritation in rabbits after single- and multiple-dose applications. Additionally, ex vivo penetration assays on isolated porcine cornea and pharmacokinetics analyses in various rabbit eye compartments demonstrated the superiority of the NCs in retention and permeation into the anterior chamber of the eye compared to the free drug dissolved in oil. Moreover, multiple dose ocular instillation of the NCs in rats allowed high tacrolimus levels in the eye with very low plasma concentrations. Finally, the developed delivery system achieved a significant decrease in four typical inflammatory markers in a murine model of keratitis, an anterior chamber inflammation. Furthermore, these NCs, applied as eye drops, displayed clinical and histological efficacy in the mainly posterior chamber inflammation model of murine, experimental auto-immune uveitis.


Assuntos
Nanocápsulas , Uveíte , Animais , Inflamação/tratamento farmacológico , Camundongos , Nanocápsulas/uso terapêutico , Soluções Oftálmicas/uso terapêutico , Coelhos , Ratos , Suínos , Tacrolimo , Uveíte/tratamento farmacológico
6.
Blood Adv ; 4(9): 2049-2057, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32396613

RESUMO

Membrane attack complexes (MACs; C5b-9) assembled after complement activation can directly injure self-tissues, leading to various diseases. Eculizumab, a monoclonal antibody (mAb) against complement component C5, is being used in the clinic to treat diseases in which MAC-mediated tissue damage is a primary cause. However, C5 is not a selective target for MAC assembly inhibition, and some patients respond incompletely or not at all to the eculizumab treatment. Therefore, C6, the next essential component in the terminal pathway of complement activation, may be an alternative target for the selective inhibition of MAC formation. Surprisingly, few reports describe a functional blockade of C6 using a specific mAb. Here, we report the development of an anti-human C6 mAb (clone 1C9) that recognizes C6 both in free circulation and within C5b6 complexes. This mAb blocked C7 binding to C5b6 complexes and consequently inhibited MAC formation and protected affected paroxysmal nocturnal hemoglobinuria patient red blood cells from MAC-mediated damage in vitro. In addition, this mAb cross-reacts with rhesus monkey but not mouse complement C6. Finally, 1C9 significantly reduced human complement-mediated intravascular hemolysis in vivo in a mouse model. These results suggest that the anti-C6 mAb holds promise as a new therapeutic agent that selectively targets MAC for many complement-mediated pathological conditions.


Assuntos
Complemento C6 , Complexo de Ataque à Membrana do Sistema Complemento , Animais , Ativação do Complemento , Complemento C5 , Hemólise , Humanos , Camundongos
7.
Infect Immun ; 77(6): 2392-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19349424

RESUMO

Our previous studies showed that surfactant protein D (SP-D) is present in human tear fluid and that it can protect corneal epithelial cells against bacterial invasion. Here we developed a novel null-infection model to test the hypothesis that SP-D contributes to the clearance of viable Pseudomonas aeruginosa from the healthy ocular surface in vivo. Healthy corneas of Black Swiss mice were inoculated with 10(7) or 10(9) CFU of invasive (PAO1) or cytotoxic (6206) P. aeruginosa. Viable counts were performed on tear fluid collected at time points ranging from 3 to 14 h postinoculation. Healthy ocular surfaces cleared both P. aeruginosa strains efficiently, even when 10(9) CFU was used: e.g., <0.01% of the original inoculum was recoverable after 3 h. Preexposure of eyes to bacteria did not enhance clearance. Clearance of strain 6206 (low protease producer), but not strain PAO1 (high protease producer), was delayed in SP-D gene-targeted (SP-D(-/-)) knockout mice. A protease mutant of PAO1 (PAO1 lasA lasB aprA) was cleared more efficiently than wild-type PAO1, but this difference was negligible in SP-D(-/-) mice, which were less able to clear the protease mutant. Experiments to study mechanisms for these differences revealed that purified elastase could degrade tear fluid SP-D in vivo. Together, these data show that SP-D can contribute to the clearance of P. aeruginosa from the healthy ocular surface and that proteases can compromise that clearance. The data also suggest that SP-D degradation in vivo is a mechanism by which P. aeruginosa proteases could contribute to virulence.


Assuntos
Proteínas de Bactérias/metabolismo , Córnea/imunologia , Córnea/microbiologia , Elastase Pancreática/metabolismo , Pseudomonas aeruginosa/patogenicidade , Proteína D Associada a Surfactante Pulmonar/antagonistas & inibidores , Proteína D Associada a Surfactante Pulmonar/imunologia , Animais , Contagem de Colônia Microbiana , Córnea/patologia , Camundongos , Camundongos Knockout , Proteína D Associada a Surfactante Pulmonar/deficiência , Lágrimas/microbiologia
8.
Ocul Surf ; 17(1): 119-133, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30439473

RESUMO

PURPOSE: Contact lens wear carries a risk of complications, including corneal infection. Solving these complications has been hindered by limitations of existing animal models. Here, we report development of a new murine model of contact lens wear. METHODS: C57BL/6 mice were fitted with custom-made silicone-hydrogel contact lenses with or without prior inoculation with Pseudomonas aeruginosa (PAO1-GFP). Contralateral eyes served as controls. Corneas were monitored for pathology, and examined ex vivo using high-magnification, time-lapse imaging. Fluorescent reporter mice allowed visualization of host cell membranes and immune cells. Lens-colonizing bacteria were detected by viable counts and FISH. Direct-colony PCR was used for bacterial identification. RESULTS: Without deliberate inoculation, lens-wearing corneas remained free of visible pathology, and retained a clarity similar to non-lens wearing controls. CD11c-YFP reporter mice revealed altered numbers, and distribution, of CD11c-positive cells in lens-wearing corneas after 24 h. Worn lenses showed bacterial colonization, primarily by known conjunctival or skin commensals. Corneal epithelial cells showed vacuolization during lens wear, and after 5 days, cells with phagocyte morphology appeared in the stroma that actively migrated over resident keratocytes that showed altered morphology. Immunofluorescence confirmed stromal Ly6G-positive cells after 5 days of lens wear, but not in MyD88 or IL-1R gene-knockout mice. P. aeruginosa-contaminated lenses caused infectious pathology in most mice from 1 to 13 days. CONCLUSIONS: This murine model of contact lens wear appears to faithfully mimic events occurring during human lens wear, and could be valuable for experiments, not possible in humans, that help solve the pathogenesis of lens-related complications.


Assuntos
Lentes de Contato , Córnea/microbiologia , Infecções Oculares Bacterianas/microbiologia , Ceratite/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/isolamento & purificação , Receptores Tipo I de Interleucina-1/genética , Animais , Contagem de Colônia Microbiana , Lentes de Contato/efeitos adversos , Córnea/patologia , Modelos Animais de Doenças , Infecções Oculares Bacterianas/metabolismo , Infecções Oculares Bacterianas/patologia , Ceratite/metabolismo , Ceratite/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/patologia , Receptores Tipo I de Interleucina-1/metabolismo , Tomografia de Coerência Óptica
9.
FEMS Immunol Med Microbiol ; 54(2): 177-84, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18657106

RESUMO

We reported previously that surfactant protein D (SP-D) was present in human tears and corneal epithelial cells, and that it contributed to tear fluid protection of those cells against Pseudomonas aeruginosa invasion. This suggested a role in ocular innate immunity. Here, we explored the effects of bacterial challenge on SP-D expression by human corneal epithelial cells. Results showed that these cells produced and secreted SP-D constitutively in culture, and that production (mRNA, protein) and secretion of SP-D were upregulated after exposure to heat-killed P. aeruginosa or to purified flagellin or lipopolysaccharide. To begin exploring the mechanism for flagellin-mediated SP-D induction, cells were exposed to purified flagellin or flagellin mutated in the TLR-5-binding domain (L94A, L88A) which reduces IL-8 secretion by A549 respiratory cells. Mutated flagellin did not upregulate IL-8 expression in corneal epithelial cells, but did induce SP-D responses. Mitogen-activated protein kinase inhibitors, especially the JNK inhibitor SP600125, reduced secretion of SP-D, but not production, in the presence of P. aeruginosa. These data show that while SP-D and IL-8 corneal responses are each induced by P. aeruginosa or its antigens, they can involve different regions of the same ligand. The data suggest that separate mechanisms may regulate SP-D secretion and production by human corneal epithelia.


Assuntos
Epitélio Corneano/imunologia , Epitélio Corneano/metabolismo , Pseudomonas aeruginosa/imunologia , Proteína D Associada a Surfactante Pulmonar/metabolismo , Análise de Variância , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Linhagem Celular Transformada , Inibidores Enzimáticos/farmacologia , Epitélio Corneano/microbiologia , Flagelina/genética , Flagelina/imunologia , Regulação da Expressão Gênica , Humanos , Interleucina-8/metabolismo , Ligantes , Lipopolissacarídeos/imunologia , Mutação Puntual , Pseudomonas aeruginosa/genética , Proteína D Associada a Surfactante Pulmonar/genética , Regulação para Cima
10.
J Cell Biol ; 217(2): 731-744, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29191848

RESUMO

Skin and mucosal epithelia deploy antimicrobial peptides (AMPs) to eliminate harmful microbes. We reported that the intermediate filament keratin 6a (K6a) is constitutively processed into antimicrobial fragments in corneal epithelial cells. In this study, we show that K6a network remodeling is a host defense response that directly up-regulates production of keratin-derived AMPs (KAMPs) by the ubiquitin-proteasome system (UPS). Bacterial ligands trigger K6a phosphorylation at S19, S22, S37, and S60, leading to network disassembly. Mutagenic analysis of K6a confirmed that the site-specific phosphorylation augmented its solubility. K6a in the cytosol is ubiquitinated by cullin-RING E3 ligases for subsequent proteasomal processing. Without an appreciable increase in K6a gene expression and proteasome activity, a higher level of cytosolic K6a results in enhanced KAMP production. Although proteasome-mediated proteolysis is known to produce antigenic peptides in adaptive immunity, our findings demonstrate its new role in producing AMPs for innate immune defense. Manipulating K6a phosphorylation or UPS activity may provide opportunities to harness the innate immunity of epithelia against infection.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Queratina-6/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/química , Células Cultivadas , Epitélio Corneano/citologia , Epitélio Corneano/metabolismo , Humanos , Testes de Sensibilidade Microbiana
11.
Ocul Surf ; 15(1): 88-96, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27670247

RESUMO

PURPOSE: The type III secretion system (T3SS) is a significant virulence determinant for Pseudomonas aeruginosa. Using a rodent model, we found that contact lens (CL)-related corneal infections were associated with lens surface biofilms. Here, we studied the impact of human tear fluid on CL-associated biofilm growth and T3SS expression. METHODS: P. aeruginosa biofilms were formed on contact lenses for up to 7 days with or without human tear fluid, then exposed to tear fluid for 5 or 24 h. Biofilms were imaged using confocal microscopy. Bacterial culturability was quantified by viable counts, and T3SS gene expression measured by RT-qPCR. Controls included trypticase soy broth, PBS and planktonic bacteria. RESULTS: With or without tear fluid, biofilms grew to ∼108 CFU viable bacteria by 24 h. Exposing biofilms to tear fluid after they had formed without it on lenses reduced bacterial culturability ∼180-fold (P<.001). CL growth increased T3SS gene expression versus planktonic bacteria [5.46 ± 0.24-fold for T3SS transcriptional activitor exsA (P=.02), and 3.76 ± 0.36-fold for T3SS effector toxin exoS (P=.01)]. Tear fluid further enhanced exsA and exoS expression in CL-grown biofilms, but not planktonic bacteria, by 2.09 ± 0.38-fold (P=.04) and 1.89 ± 0.26-fold (P<.001), respectively. CONCLUSIONS: Considering the pivitol role of the T3SS in P. aeruginosa infections, its induction in CL-grown P. aeruginosa biofilms by tear fluid might contribute to the pathogenesis of CL-related P. aeruginosa keratitis.


Assuntos
Lentes de Contato , Biofilmes , Humanos , Pseudomonas aeruginosa , Sistemas de Secreção Tipo III , Virulência
12.
Sci Rep ; 7(1): 13829, 2017 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-29062042

RESUMO

Previously we reported that corneal epithelial barrier function against Pseudomonas aeruginosa was MyD88-dependent. Here, we explored contributions of MyD88-dependent receptors using vital mouse eyes and confocal imaging. Uninjured IL-1R (-/-) or TLR4 (-/-) corneas, but not TLR2 (-/-), TLR5 (-/-), TLR7 (-/-), or TLR9 (-/-), were more susceptible to P. aeruginosa adhesion than wild-type (3.8-fold, 3.6-fold respectively). Bacteria adherent to the corneas of IL-1R (-/-) or TLR5 (-/-) mice penetrated beyond the epithelial surface only if the cornea was superficially-injured. Bone marrow chimeras showed that bone marrow-derived cells contributed to IL-1R-dependent barrier function. In vivo, but not ex vivo, stromal CD11c+ cells responded to bacterial challenge even when corneas were uninjured. These cells extended processes toward the epithelial surface, and co-localized with adherent bacteria in superficially-injured corneas. While CD11c+ cell depletion reduced IL-6, IL-1ß, CXCL1, CXCL2 and CXCL10 transcriptional responses to bacteria, and increased susceptibility to bacterial adhesion (>3-fold), the epithelium remained resistant to bacterial penetration. IL-1R (-/-) corneas also showed down-regulation of IL-6 and CXCL1 genes with and without bacterial challenge. These data show complex roles for TLR4, TLR5, IL-1R and CD11c+ cells in constitutive epithelial barrier function against P. aeruginosa, with details dependent upon in vivo conditions.


Assuntos
Antígeno CD11c/imunologia , Permeabilidade da Membrana Celular , Epitélio Corneano/imunologia , Regulação da Expressão Gênica , Fator 88 de Diferenciação Mieloide/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Animais , Aderência Bacteriana , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/microbiologia , Antígeno CD11c/metabolismo , Células Cultivadas , Epitélio Corneano/metabolismo , Epitélio Corneano/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/microbiologia , Receptores de Interleucina-1/fisiologia , Transdução de Sinais , Receptor 4 Toll-Like/fisiologia , Receptor 5 Toll-Like/fisiologia
13.
Front Microbiol ; 7: 1799, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27891122

RESUMO

Antibiotic resistance is a pressing global health problem that threatens millions of lives each year. Natural antimicrobial peptides and their synthetic derivatives, including peptoids and peptidomimetics, are promising candidates as novel antibiotics. Recently, the C-terminal glycine-rich fragments of human epithelial keratin 6A were found to have bactericidal and cytoprotective activities. Here, we used an improved 2-dimensional NMR method coupled with a new protocol for structural refinement by low temperature simulated annealing to characterize the solution structure of these kerain-derived antimicrobial peptides (KAMPs). Two specific KAMPs in complex with membrane mimicking sodium dodecyl sulfate (SDS) micelles displayed amphipathic conformations with only local bends and turns, and a central 10-residue glycine-rich hydrophobic strip that is central to bactericidal activity. To our knowledge, this is the first report of non-αß structure for human antimicrobial peptides. Direct observation of Staphylococcus aureus and Pseudomonas aeruginosa by scanning and transmission electron microscopy showed that KAMPs deformed bacterial cell envelopes and induced pore formation. Notably, in competitive binding experiments, KAMPs demonstrated binding affinities to LPS and LTA that did not correlate with their bactericidal activities, suggesting peptide-LPS and peptide-LTA interactions are less important in their mechanisms of action. Moreover, immunoprecipitation of KAMPs-bacterial factor complexes indicated that membrane surface lipoprotein SlyB and intracellular machineries NQR sodium pump and ribosomes are potential molecular targets for the peptides. Results of this study improve our understanding of the bactericidal function of epithelial cytokeratin fragments, and highlight an unexplored class of human antimicrobial peptides, which may serve as non-αß peptide scaffolds for the design of novel peptide-based antibiotics.

14.
PLoS One ; 8(2): e57850, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469087

RESUMO

Mucosal surfaces regulate defenses against infection and excessive inflammation. We previously showed that human tears upregulated epithelial expression of genes encoding RNase7 and ST2, which inhibited Pseudomonas aeruginosa invasion of human corneal epithelial cells. Here, microRNA microarrays were used to show that a combination of tear fluid exposure (16 h) then P. aeruginosa antigens (3 h) upregulated miR-762 and miR-1207, and down-regulated miR-92 and let-7b (all > 2-fold) in human corneal epithelial cells compared to P. aeruginosa antigens alone. RT-PCR confirmed miR-762 upregulation ∼ 3-fold in tear-antigen exposed cells. Without tears or antigens, an antagomir reduced miR-762 expression relative to scrambled controls by ∼50%, increased expression of genes encoding RNase7 (∼80 %), ST2 (∼58%) and Rab5a (∼75%), without affecting P. aeruginosa internalization. However, P. aeruginosa invasion was increased > 3-fold by a miR-762 mimic which reduced RNase7 and ST2 gene expression. Tear fluid alone also induced miR-762 expression ∼ 4-fold, which was reduced by the miR-762 antagomir. Combination of tear fluid and miR-762 antagomir increased RNase7 and ST2 gene expression. These data show that mucosal fluids, such as tears, can modulate epithelial microRNA expression to regulate innate defense genes, and that miR-762 negatively regulates RNase7, ST2 and Rab5a genes. Since RNase7 and ST2 inhibit P. aeruginosa internalization, and are upregulated by tear fluid, other tear-induced mechanisms must counteract inhibitory effects of miR-762 to regulate resistance to bacteria. These data also suggest a complex relationship between tear induction of miR-762, its modulation of innate defense genes, and P. aeruginosa internalization.


Assuntos
Epitélio Corneano/citologia , MicroRNAs/genética , Pseudomonas aeruginosa/imunologia , Receptores de Superfície Celular/genética , Ribonucleases/genética , Lágrimas/metabolismo , Regulação para Cima/imunologia , Antígenos de Bactérias/imunologia , Regulação para Baixo/imunologia , Epitélio Corneano/microbiologia , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1 , Oligonucleotídeos/genética , Pseudomonas aeruginosa/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas rab5 de Ligação ao GTP/genética
15.
JAMA Ophthalmol ; 131(2): 147-53, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23411878

RESUMO

OBJECTIVES: To determine whether cytotoxic and invasive Pseudomonas aeruginosa strains differentially influence clinical presentation, outcomes, or therapeutic response in bacterial keratitis. METHODS: Pseudomonas aeruginosa isolates from the National Eye Institute-funded Steroids for Corneal Ulcers Trial were subtyped as cytotoxic or invasive strains. The main outcome measure compared between the 2 subtypes was change in visual acuity at 3 months using Huber robust regression, adjusting for topical corticosteroid treatment. RESULTS: Of 101 confirmed P aeruginosa isolates from the Steroids for Corneal Ulcers Trial, 74 had a classically cytotoxic or invasive genotype. While corneal ulcers caused by genotypically invasive P aeruginosa strains were associated at presentation with significantly better visual acuity than corneal ulcers caused by genotypically cytotoxic P aeruginosa strains when adjusting for the effect of ulcer location (P= .008), invasive ulcers had improved significantly less than cytotoxic ulcers at 3 months (0.35; 95% CI, 0.04-0.66 logMAR; P= .03 [3.5-line difference]). Compared with topical moxifloxacin alone, adjunctive treatment with topical corticosteroids was associated with significantly more improvement in visual acuity in the invasive subgroup (P= .04) but was associated with less improvement in visual acuity in the cytotoxic subgroup (P= .07). CONCLUSIONS: Rational profiling of differentially expressed virulence determinants (eg, cytotoxicity and invasiveness for P aeruginosa) could be used as a tool for decision making in the management of infections to optimize outcomes.


Assuntos
Úlcera da Córnea/microbiologia , Infecções Oculares Bacterianas/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Anti-Infecciosos/uso terapêutico , Compostos Aza/uso terapêutico , Toxinas Bacterianas/genética , Técnicas de Tipagem Bacteriana , Úlcera da Córnea/diagnóstico , Úlcera da Córnea/tratamento farmacológico , DNA Bacteriano/análise , Exotoxinas/fisiologia , Infecções Oculares Bacterianas/diagnóstico , Infecções Oculares Bacterianas/tratamento farmacológico , Fluoroquinolonas , Genótipo , Glucocorticoides/uso terapêutico , Humanos , L-Lactato Desidrogenase/metabolismo , Pessoa de Meia-Idade , Moxifloxacina , Reação em Cadeia da Polimerase , Infecções por Pseudomonas/diagnóstico , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/isolamento & purificação , Pseudomonas aeruginosa/fisiologia , Quinolinas/uso terapêutico , Virulência/fisiologia , Acuidade Visual/fisiologia
16.
J Clin Invest ; 122(10): 3665-77, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23006328

RESUMO

Epithelial cells express antimicrobial proteins in response to invading pathogens, although little is known regarding epithelial defense mechanisms during healthy conditions. Here we report that epithelial cytokeratins have innate defense properties because they constitutively produce cytoprotective antimicrobial peptides. Glycine-rich C-terminal fragments derived from human cytokeratin 6A were identified in bactericidal lysate fractions of human corneal epithelial cells. Structural analysis revealed that these keratin-derived antimicrobial peptides (KDAMPs) exhibited coil structures with low α-helical content. Synthetic analogs of these KDAMPS showed rapid bactericidal activity against multiple pathogens and protected epithelial cells against bacterial virulence mechanisms, while a scrambled peptide showed no bactericidal activity. However, the bactericidal activity of a specific KDAMP was somewhat reduced by glycine-alanine substitutions. KDAMP activity involved bacterial binding and permeabilization, but the activity was unaffected by peptide charge or physiological salt concentration. Knockdown of cytokeratin 6A markedly reduced the bactericidal activity of epithelial cell lysates in vitro and increased the susceptibility of murine corneas to bacterial adherence in vivo. These data suggest that epithelial cytokeratins function as endogenous antimicrobial peptides in the host defense against infection and that keratin-derived antimicrobials may serve as effective therapeutic agents.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Células Epiteliais/imunologia , Epitélio Corneano/imunologia , Queratina-6/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/isolamento & purificação , Bactérias/efeitos dos fármacos , Bactérias/imunologia , Bactérias/ultraestrutura , Aderência Bacteriana/efeitos dos fármacos , Fracionamento Celular , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Conjuntivite Bacteriana/microbiologia , Conjuntivite Bacteriana/prevenção & controle , Epitélio Corneano/citologia , Humanos , Imunidade Inata , Queratina-6/química , Queratina-6/genética , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/fisiologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Relação Estrutura-Atividade
17.
Invest Ophthalmol Vis Sci ; 53(1): 448-53, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22167094

RESUMO

PURPOSE: Pseudomonas aeruginosa isolates from microbial keratitis are invasive or cytotoxic toward mammalian cells, depending on their type III secreted toxins. Cytotoxic strains express ExoU, a phospholipase that contributes to corneal virulence. This study determined whether the ExoU phospholipase domain is required for P. aeruginosa traversal of the human corneal epithelium. METHODS: P. aeruginosa traversal of airlifted, multilayered, human corneal epithelial cells was quantified in vitro up to 8 hours after apical inoculation with ∼106 cfu of strain PA14, or an isogenic exoU mutant (PA14ΔexoU). In addition, PA14ΔexoU or its triple effector mutant PA14ΔexoUΔexoTΔexoY, were complemented with exoU (pUCPexoU), phospholipase-inactive exoU (pUCPexoUD344A), or control plasmid (pUCP18). Transepithelial resistance (TER) was measured (by epithelial volt ohmmeter), and cytotoxicity was determined by trypan blue staining. RESULTS: PA14 traversed more efficiently than its exoU mutant at 4, 6, and 8 hours after inoculation (100-, 20-, and 8-fold, respectively; P < 0.05), but not at 2 hours. Cells exposed to PA14 lost TER to baseline (P < 0.05). Controls confirmed PA14 cytotoxicity toward these corneal epithelial cells that was absent with exoU mutants. Epithelial traversal, cytotoxicity, and lost TER were restored for PA14ΔexoU, or PA14ΔexoUΔexoTΔexoY, by complementation with pUCPexoU, but not by complementation with pUCPexoUD344A. CONCLUSIONS: Traversal of multilayered corneal epithelia in vitro by cytotoxic P. aeruginosa requires ExoU with an active phospholipase domain. Correlative loss of TER with traversal by wild-type, or exoU-complemented, bacteria suggests involvement of epithelial cell death and/or lost tight junction integrity. However, traversal by exoU mutants without reduced TER suggests that additional mechanisms are also operative.


Assuntos
Proteínas de Bactérias/metabolismo , Epitélio Corneano/microbiologia , Fosfolipases/metabolismo , Pseudomonas aeruginosa/fisiologia , Células Cultivadas , Impedância Elétrica , Epitélio Corneano/metabolismo , Humanos , Estrutura Terciária de Proteína
18.
Sci Rep ; 1: 8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22355527

RESUMO

Mucosal epithelial cells, including those at the ocular surface, resist infection by most microbes in vivo but can be susceptible to microbial virulence in vitro. While fluids bathing mucosal surfaces (e.g. tears) contain antimicrobials, potentially pathogenic microbes often thrive in these fluids, suggesting that additional mechanisms mediate epithelial resistance in vivo. Here, tear fluid acted directly upon epithelial cells to enhance their resistance to bacterial invasion and cytotoxicity. Resistance correlated with tear fluid-magnified activation of NFκB and AP-1 transcription factors in epithelial cells in response to bacterial antigens, suggesting priming of innate defense pathways. Further analysis revealed differential regulation of potential epithelial cell defense genes by tears. siRNA knockdown confirmed involvement of at least two factors, RNase7 and ST-2, for which tears increased mRNA levels, in protection against bacterial invasion. Thus, the role of mucosal fluids in defense can include modulation of epithelial immunity, in addition to direct effects on microbes.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/fisiologia , Imunidade nas Mucosas/imunologia , Pseudomonas aeruginosa/fisiologia , Lágrimas/imunologia , Lágrimas/microbiologia , Células Cultivadas , Humanos
19.
PLoS One ; 6(8): e24008, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21901151

RESUMO

While a plethora of in vivo models exist for studying infectious disease and its resolution, few enable factors involved in the maintenance of health to be studied in situ. This is due in part to a paucity of tools for studying subtleties of bacterial-host interactions at a cellular level within live organs or tissues, requiring investigators to rely on overt outcomes (e.g. pathology) in their research. Here, a suite of imaging technologies were combined to enable 3D and temporal subcellular localization and quantification of bacterial distribution within the murine cornea without the need for tissue processing or dissection. These methods were then used to demonstrate the importance of MyD88, a central adaptor protein for Toll-Like Receptor (TLR) mediated signaling, in protecting a multilayered epithelium against both adhesion and traversal by the opportunistic bacterial pathogen Pseudomonas aeruginosa ex vivo and in vivo.


Assuntos
Aderência Bacteriana/fisiologia , Córnea/microbiologia , Fator 88 de Diferenciação Mieloide/metabolismo , Animais , Córnea/metabolismo , Feminino , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos CBA , Camundongos Knockout , Pseudomonas aeruginosa/fisiologia , Transdução de Sinais/fisiologia , Receptores Toll-Like/metabolismo
20.
Invest Ophthalmol Vis Sci ; 52(3): 1368-77, 2011 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-21051692

RESUMO

PURPOSE: Mechanisms determining epithelial resistance versus susceptibility to microbial traversal in vivo remain poorly understood. Here, a novel murine model was used to explore factors influencing the corneal epithelial barrier to Pseudomonas aeruginosa penetration. METHODS: Murine corneas were blotted with tissue paper before inoculation with green fluorescent protein-expressing P. aeruginosa. The impact of blotting on epithelial integrity was evaluated by susceptibility to fluorescein staining and histology. Using fluorescence imaging, blotted corneas were compared to nonblotted corneas for susceptibility to bacterial binding and epithelial penetration after 5 hours or were monitored for disease development. In some experiments, inoculation was performed ex vivo to exclude tear fluid or corneas were pretreated with EGTA to disrupt Ca(2+)-dependent factors. The role of surfactant protein D (SP-D), which inhibits P. aeruginosa cell invasion in vitro, was examined using knockout mice. RESULTS: Blotting enabled fluorescein penetration through the epithelium into the underlying stroma without obvious disruption to corneal morphology. Although blotting enabled bacterial binding to the otherwise adhesion-resistant epithelial surface, adherent bacteria did not penetrate the surface or initiate pathology. In contrast, bacteria penetrated blotted corneas after EGTA treatment and in SP-D knockouts. Visible disease occurred and progressed only in aged, blotted, and EGTA-treated, SP-D knockout mice. CONCLUSIONS: Neither fluorescein staining nor bacterial adhesion necessarily predict or enable corneal susceptibility to bacterial penetration or disease. Corneal epithelial defenses limiting traversal by adherent bacteria include EGTA-sensitive factors and SP-D. Understanding mechanisms modulating epithelial traversal by microbes could improve our understanding of susceptibility to infection and may indicate new strategies for preventing disease.


Assuntos
Aderência Bacteriana/fisiologia , Úlcera da Córnea/microbiologia , Epitélio Corneano/fisiologia , Infecções Oculares Bacterianas/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/fisiologia , Junções Íntimas/fisiologia , Animais , Membrana Basal/fisiologia , Contagem de Colônia Microbiana , Ácido Egtázico/farmacologia , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/microbiologia , Proteínas da Matriz Extracelular/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Fluorofotometria , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína D Associada a Surfactante Pulmonar/fisiologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA