Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Infect Immun ; 82(2): 851-63, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478098

RESUMO

Klebsiella pneumoniae has become an important pathogen in recent years. Although most cases of K. pneumoniae endogenous endophthalmitis occur via hematogenous spread, it is not yet clear which microbial and host factors are responsible for the ability of K. pneumoniae to cross the blood-retinal barrier (BRB). In the present study, we show that in an in vitro model of BRB based on coculturing primary bovine retinal endothelial cells (BREC) and primary bovine retinal pericytes (BRPC), K. pneumoniae infection determines changes of transendothelial electrical resistance (TEER) and permeability to sodium fluorescein. In the coculture model, bacteria are able to stimulate the enzyme activities of endothelial cytosolic and Ca(2+)-independent phospholipase A2s (cPLA2 and iPLA2). These results were confirmed by the incremental expression of cPLA2, iPLA2, cyclo-oxygenase-1 (COX1), and COX2 in BREC, as well as by cPLA2 phosphorylation. In supernatants of K. pneumoniae-stimulated cocultures, increases in prostaglandin E2 (PGE2), interleukin-6 (IL-6), IL-8, and vascular endothelial growth factor (VEGF) production were found. Incubation with K. pneumoniae in the presence of arachidonoyl trifluoromethyl ketone (AACOCF3) or bromoenol lactone (BEL) caused decreased PGE2 and VEGF release. Scanning electron microscopy and transmission electron microscopy images of BREC and BRPC showed adhesion of K. pneumoniae to the cells, but no invasion occurred. K. pneumoniae infection also produced reductions in pericyte numbers; transfection of BREC cocultured with BRPC and of human retinal endothelial cells (HREC) cocultured with human retinal pericytes (HRPC) with small interfering RNAs (siRNAs) targeted to cPLA2 and iPLA2 restored the pericyte numbers and the TEER and permeability values. Our results show the proinflammatory effect of K. pneumoniae on BREC, suggest a possible mechanism by which BREC and BRPC react to the K. pneumoniae infection, and may provide physicians and patients with new ways of fighting blinding diseases.


Assuntos
Barreira Hematorretiniana/microbiologia , Barreira Hematorretiniana/patologia , Células Endoteliais/microbiologia , Inflamação/microbiologia , Inflamação/fisiopatologia , Klebsiella pneumoniae/imunologia , Pericitos/microbiologia , Animais , Bovinos , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/fisiologia , Perfilação da Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Klebsiella pneumoniae/patogenicidade , Pericitos/fisiologia , Permeabilidade
2.
Microbiol Spectr ; 10(4): e0165822, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-35913202

RESUMO

Candida albicans is the leading cause of endogenous fungal endophthalmitis; however, its pathobiology studies are limited. Moreover, the contribution of host factors in the pathogenesis of Candida endophthalmitis remains unclear. In the present study, we developed a murine model of C. albicans endogenous endophthalmitis and investigated the molecular pathobiology of ocular candidiasis and blood-retinal barrier permeability. Our data show that intravenous injection of C. albicans in immunocompetent C57BL/6 mice led to endogenous endophthalmitis without causing mortality, and C. albicans was detected in the eyes at 3 days postinfection and persisted for up to 10 days. The intraocular presence of C. albicans coincided with a decrease in retinal function and increased expression of inflammatory mediators (tumor necrosis factor alpha [TNF-α], interleukin 1ß [IL-1ß], MIP2, and KC) and antimicrobial peptides (human ß-defensins [hBDs] and LL37) in mouse retinal tissue. C. albicans infection disrupted the blood-retinal barrier (BRB) by decreasing the expression of tight junction (ZO-1) and adherens junction (E-cadherin, N/R-cadherin) proteins. In vitro studies using human retinal pigment epithelial (ARPE-19) cells showed time-dependent activation of eIF2α, extracellular signal-related kinase (ERK), and NF-κB signaling and decreased activity of AMP-activated protein kinase (AMPK) leading to the induction of an inflammatory response upon C. albicans infection. Moreover, C. albicans-infected cells exhibited increased cellular permeability coinciding with a reduction in cellular junction proteins. Overall, our study provides new insight into the molecular pathogenesis of C. albicans endogenous endophthalmitis. Furthermore, the experimental models developed in the study can be used to identify newer therapeutic targets or test the efficacy of drugs to treat and prevent fungal endophthalmitis. IMPORTANCE Patients with candidemia often experience endophthalmitis, a blinding infectious eye disease. However, the pathogenesis of Candida endophthalmitis is not well understood. Here, using in vivo and in vitro experimental models, we describe events leading to the invasion of Candida into the eye. We show that Candida from the systemic circulation disrupts the protective blood-retinal barrier and causes endogenous endophthalmitis. Our study highlights an important role of retinal pigment epithelial cells in evoking innate inflammatory and antimicrobial responses toward C. albicans infection. This study allows a better understanding of the pathobiology of fungal endophthalmitis, which can lead to the discovery of novel therapeutic targets to treat ocular fungal infections.


Assuntos
Candidíase , Endoftalmite , Infecções Oculares Fúngicas , Animais , Barreira Hematorretiniana/microbiologia , Barreira Hematorretiniana/patologia , Candida , Candida albicans , Endoftalmite/tratamento farmacológico , Endoftalmite/microbiologia , Endoftalmite/patologia , Infecções Oculares Fúngicas/tratamento farmacológico , Infecções Oculares Fúngicas/microbiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pigmentos da Retina/uso terapêutico
3.
Prog Brain Res ; 256(1): 79-97, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32958216

RESUMO

Over the last decade, new evidence has become increasingly more compelling that commensal microflora profoundly influences the maturation and function of resident immune cells in host physiology. The concept of gut-retina axis is actively being explored. Studies have revealed a critical role of commensal microbes linked with neuronal stress, immune responses, and neurodegeneration in the retina. Microbial dysbiosis changes the blood-retina barrier permeability and modulates T cell-mediated autoimmunity to contribute to the pathogenesis of retinal diseases, such as glaucoma. Heat shock proteins (HSPs), which are evolutionarily conserved, are thought to function both as neuroprotectant and pathogenic antigens of T cells contributing to cell protection and tissue damage, respectively. Activated microglia recruit and interact with T cells during this process. Glaucoma, characterized by the progressive loss of retinal ganglion cells, is the leading cause of irreversible blindness. With nearly 70 million people suffering glaucoma worldwide, which doubles the number of patients with Alzheimer's disease, it represents the most frequent neurodegenerative disease of the central nervous system (CNS). Thus, understanding the mechanism of neurodegeneration in glaucoma and its association with the function of commensal microflora may help unveil the secrets of many neurodegenerative disorders in the CNS and develop novel therapeutic interventions.


Assuntos
Barreira Hematorretiniana , Microbioma Gastrointestinal , Glaucoma , Proteínas de Choque Térmico , Degeneração Neural , Retina , Linfócitos T , Animais , Barreira Hematorretiniana/imunologia , Barreira Hematorretiniana/metabolismo , Barreira Hematorretiniana/microbiologia , Barreira Hematorretiniana/patologia , Glaucoma/imunologia , Glaucoma/metabolismo , Glaucoma/microbiologia , Glaucoma/patologia , Proteínas de Choque Térmico/imunologia , Proteínas de Choque Térmico/metabolismo , Humanos , Degeneração Neural/imunologia , Degeneração Neural/metabolismo , Degeneração Neural/microbiologia , Degeneração Neural/patologia , Retina/imunologia , Retina/metabolismo , Retina/microbiologia , Retina/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
4.
Infect Immun ; 76(4): 1358-67, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18268029

RESUMO

Most Bacillus cereus toxin production is controlled by the quorum-sensing-dependent, pleiotropic global regulator plcR, which contributes to the organism's virulence in the eye. The purpose of this study was to analyze the effects of B. cereus infection and plcR-regulated toxins on the barrier function of retinal pigment epithelium (RPE) cells, the primary cells of the blood-retina barrier. Human ARPE-19 cells were apically inoculated with wild-type or quorum-sensing-deficient B. cereus, and cytotoxicity was analyzed. plcR-regulated toxins were not required for B. cereus-induced RPE cytotoxicity, but these toxins did increase the rate of cell death, primarily by necrosis. B. cereus infection of polarized RPE cell monolayers resulted in increased barrier permeability, independent of plcR-regulated toxins. Loss of both occludin and ZO-1 expression occurred by 8 h postinfection, but alterations in tight junctions appeared to precede cytotoxicity. Of the several proinflammatory cytokines analyzed, only interleukin-6 was produced in response to B. cereus infection. These results demonstrate the deleterious effects of B. cereus infection on RPE barrier function and suggest that plcR-regulated toxins may not contribute significantly to RPE barrier permeability during infection.


Assuntos
Bacillus cereus/fisiologia , Barreira Hematorretiniana/microbiologia , Barreira Hematorretiniana/patologia , Linhagem Celular , Humanos , Permeabilidade , Transporte Proteico , ATPase Trocadora de Sódio-Potássio/metabolismo , Junções Íntimas/metabolismo
5.
PLoS One ; 13(3): e0194982, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29584775

RESUMO

Ocular tuberculosis (TB) commonly causes severe inflammation and vision loss in TB-endemic countries. The mechanism by which tuberculous infection becomes established in the eye is poorly understood. We have developed the zebrafish larva infected with Mycobacterium marinum as a model to study the early pathogenesis of ocular TB. We find that hematogenous bacterial seeding of the eye occurs despite a functional blood retinal barrier. Prototypical early granulomas form in response to bacteria in the eye. These granulomas involve the retinal vasculature and retinal pigment epithelium-choroid complex which are characteristic locations for human ocular TB. We find that peripheral blood monocytes are recruited to the nascent ocular granuloma further suggesting that the immune privileged nature of the eye is breached by this inflammatory focus.


Assuntos
Tuberculose Ocular/patologia , Peixe-Zebra/microbiologia , Animais , Barreira Hematorretiniana/microbiologia , Modelos Animais de Doenças , Granuloma/etiologia , Granuloma/imunologia , Larva/microbiologia , Microscopia Confocal , Monócitos/citologia , Monócitos/imunologia , Mycobacterium marinum/patogenicidade , Tecido Parenquimatoso/patologia , Tuberculose Ocular/metabolismo , Peixe-Zebra/crescimento & desenvolvimento
6.
PLoS One ; 11(5): e0154560, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27195776

RESUMO

The blood-retinal barrier (BRB) functions to maintain the immune privilege of the eye, which is necessary for normal vision. The outer BRB is formed by tightly-associated retinal pigment epithelial (RPE) cells which limit transport within the retinal environment, maintaining retinal function and viability. Retinal microvascular complications and RPE dysfunction resulting from diabetes and diabetic retinopathy cause permeability changes in the BRB that compromise barrier function. Diabetes is the major predisposing condition underlying endogenous bacterial endophthalmitis (EBE), a blinding intraocular infection resulting from bacterial invasion of the eye from the bloodstream. However, significant numbers of EBE cases occur in non-diabetics. In this work, we hypothesized that dysfunction of the outer BRB may be associated with EBE development. To disrupt the RPE component of the outer BRB in vivo, sodium iodate (NaIO3) was administered to C57BL/6J mice. NaIO3-treated and untreated mice were intravenously injected with 108 colony forming units (cfu) of Staphylococcus aureus or Klebsiella pneumoniae. At 4 and 6 days postinfection, EBE was observed in NaIO3-treated mice after infection with K. pneumoniae and S. aureus, although the incidence was higher following S. aureus infection. Invasion of the eye was observed in control mice following S. aureus infection, but not in control mice following K. pneumoniae infection. Immunohistochemistry and FITC-dextran conjugate transmigration assays of human RPE barriers after infection with an exoprotein-deficient agr/sar mutant of S. aureus suggested that S. aureus exoproteins may be required for the loss of the tight junction protein, ZO-1, and for permeability of this in vitro barrier. Our results support the clinical findings that for both pathogens, complications which result in BRB permeability increase the likelihood of bacterial transmigration from the bloodstream into the eye. For S. aureus, however, BRB permeability is not required for the development of EBE, but toxin production may facilitate EBE pathogenesis.


Assuntos
Barreira Hematorretiniana/microbiologia , Infecções Oculares Bacterianas/microbiologia , Epitélio Pigmentado da Retina/microbiologia , Angiografia , Animais , Sobrevivência Celular , Células Cultivadas , Corantes/química , Dextranos , Retinopatia Diabética/patologia , Endoftalmite/microbiologia , Azul Evans/química , Fluoresceína-5-Isotiocianato/análogos & derivados , Humanos , Imuno-Histoquímica , Iodatos/química , Klebsiella pneumoniae , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Epitélio Pigmentado da Retina/citologia , Vasos Retinianos/patologia , Staphylococcus aureus
7.
Intern Med ; 54(18): 2401-4, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26370869

RESUMO

A 67-year-old man was transported to our hospital and diagnosed with pneumococcal meningitis. We immediately administered ceftriaxone and vancomycin according to the guidelines, but did not administer dexamethasone to him because he had been previously administered antibiotics. His left eye became complicated by endogenous endophthalmitis on the next day, which resulted in blindness, although his meningitis rapidly ameliorated. In comparison to other patients who have been reported to recover from complications with endophthalmitis after the combination therapy of antibiotics, corticosteroids and vitreous surgery, we consider that this patient's poor visual outcome may have been caused by severe inflammation or the breakdown of the blood ocular barrier due to the action of S. pneumoniae. Corticosteroids may be able to successfully treat such inflammation or disruption of the blood ocular barrier.


Assuntos
Endoftalmite/microbiologia , Infecções Oculares Bacterianas/microbiologia , Meningite Pneumocócica/complicações , Idoso , Antibacterianos/uso terapêutico , Cegueira/etiologia , Barreira Hematorretiniana/microbiologia , Ceftriaxona/uso terapêutico , Quimioterapia Combinada , Endoftalmite/complicações , Infecções Oculares Bacterianas/complicações , Humanos , Masculino , Streptococcus pneumoniae , Vancomicina/uso terapêutico
8.
Invest Ophthalmol Vis Sci ; 50(8): 3783-93, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19264886

RESUMO

PURPOSE: The purpose of this study was to determine to what extent blood-retinal barrier (BRB) permeability occurred during experimental Bacillus cereus endophthalmitis and whether tight junction alterations were involved in permeability. METHODS: Mice were intravitreally injected with 100 colony-forming units of B. cereus, and eyes were analyzed at specific times after infection for permeability to fibrin and albumin, quantitation of intraocular plasma constituent leakage, production of inflammatory cytokines, and alterations in tight junction protein localization and expression at the level of the retinal pigment epithelium. RESULTS: B. cereus induced the leakage of albumin and fibrin into the aqueous and vitreous humor by 8 hours after infection. BRB permeability occurred as early as 4 hours and increased 13.30-fold compared with uninfected controls by 8 hours. Production of proinflammatory cytokines IL-6, MIP-1alpha, IL-1beta, and KC increased over the course of infection. In the retina, ZO-1 disruption began by 4 hours and was followed by decreasing occludin and ZO-1 expression at 4 and 8 hours, respectively. Tubulin condensation and RPE65 degradation occurred by 12 hours. A quorum-sensing mutant B. cereus strain caused BRB permeability comparable to that of wild-type B. cereus. Wild-type and mutant B. cereus sterile supernatants induced blood-ocular barrier permeability similarly to that of wild-type infection. CONCLUSIONS: These results indicate that BRB permeability occurs during the early stages of experimental B. cereus endophthalmitis, beginning as early as 4 hours after infection. Disruption of tight junctions at the level of the retinal pigment epithelium may contribute to barrier breakdown. Quorum-sensing dependent factors may not significantly contribute to BRB permeability.


Assuntos
Bacillus cereus/fisiologia , Barreira Hematorretiniana/microbiologia , Permeabilidade Capilar/fisiologia , Endoftalmite/microbiologia , Infecções por Bactérias Gram-Positivas/microbiologia , Animais , Humor Aquoso/metabolismo , Western Blotting , Citocinas/metabolismo , Endoftalmite/metabolismo , Endoftalmite/patologia , Fibrina/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Infecções por Bactérias Gram-Positivas/metabolismo , Infecções por Bactérias Gram-Positivas/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ocludina , Fosfoproteínas/metabolismo , Albumina Sérica/metabolismo , Junções Íntimas/metabolismo , Corpo Vítreo/metabolismo , Proteína da Zônula de Oclusão-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA