Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Intervalo de ano de publicação
2.
Pathogens ; 12(4)2023 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-37111466

RESUMO

Streptococcus agalactiae (Group B Streptococcus, GBS) is the leading cause of neonatal sepsis and meningitis but has been recently isolated from non-pregnant adults with underlying medical conditions like diabetes. Despite diabetes being a key risk factor for invasive disease, the pathological consequences during GBS infection remain poorly characterized. Here, we demonstrate the pathogenicity of the GBS90356-ST17 and COH1-ST17 strains in streptozotocin-induced diabetic mice. We show that GBS can spread through the bloodstream and colonize several tissues, presenting a higher bacterial count in diabetic-infected mice when compared to non-diabetic-infected mice. Histological sections of the lungs showed inflammatory cell infiltration, collapsed septa, and red blood cell extravasation in the diabetic-infected group. A significant increase in collagen deposition and elastic fibers were also observed in the lungs. Moreover, the diabetic group presented red blood cells that adhered to the valve wall and disorganized cardiac muscle fibers. An increased expression of KC protein, IL-1ß, genes encoding immune cell markers, and ROS (reactive oxygen species) production was observed in diabetic-infected mice, suggesting GBS promotes high levels of inflammation when compared to non-diabetic animals. Our data indicate that efforts to reverse the epidemic of diabetes could considerably reduce the incidence of invasive infection, morbidity and mortality due to GBS.

3.
Front Immunol ; 14: 1075834, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36761775

RESUMO

The inflammasomes are intracellular multimeric protein complexes consisting of an innate immune sensor, the adapter protein ASC and the inflammatory caspases-1 and/or -11 and are important for the host defense against pathogens. Activaton of the receptor leads to formation of the inflammasomes and subsequent processing and activation of caspase-1 that cleaves the proinflammatory cytokines IL-1ß and IL-18. Active caspase-1, and in some instances caspase-11, cleaves gasdermin D that translocates to the cell membrane where it forms pores resulting in the cell death program called pyroptosis. Inflammasomes can detect a range of microbial ligands through direct interaction or indirectly through diverse cellular processes including changes in ion fluxes, production of reactive oxygen species and disruption of various host cell functions. In this review, we will focus on the NLRP3, NLRP6, NLRC4 and AIM2 inflammasomes and how they are activated and regulated during infections with Gram-positive bacteria, including Staphylococcus spp., Streptococcus spp. and Listeria monocytogenes.


Assuntos
Proteínas de Transporte , Inflamassomos , Inflamassomos/metabolismo , Proteínas de Transporte/metabolismo , Caspases/metabolismo , Citocinas/metabolismo , Caspase 1/metabolismo , Bactérias Gram-Positivas/metabolismo
4.
Front Immunol ; 13: 902907, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35911737

RESUMO

Sepsis is a generalized disease characterized by an extreme response to a severe infection. Moreover, challenges remain in the diagnosis, treatment and management of septic patients. In this mini-review we demonstrate developments on cellular pathogenesis and the role of Caveolin-1 (Cav-1) in sepsis. Studies have shown that Cav-1 has a significant role in sepsis through the regulation of membrane traffic and intracellular signaling pathways. In addition, activation of apoptosis/autophagy is considered relevant for the progression and development of sepsis. However, how Cav-1 is involved in sepsis remains unclear, and the precise mechanisms need to be further investigated. Finally, the role of Cav-1 in altering cell permeability during inflammation, in sepsis caused by microorganisms, apoptosis/autophagy activation and new therapies under study are discussed in this mini-review.


Assuntos
Caveolina 1 , Sepse , Autofagia/fisiologia , Caveolina 1/genética , Caveolina 1/metabolismo , Humanos , Permeabilidade , Sepse/genética , Sepse/metabolismo , Transdução de Sinais
6.
Braz J Microbiol ; 52(1): 303-310, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33398672

RESUMO

Streptococcus agalactiae is a recognized pathogen associated with infections in neonates, elderly, and immunocompromised adults, particularly those with cancer. In the present investigation, clinical-epidemiological features, multidrug resistance profiles, and virulence genes of S. agalactiae strains isolated from cancer patients were investigated. S. agalactiae capsular distribution assays demonstrated that Ia (43.6%) and V (23.6%) types were predominantly detected among 55 clinical isolates tested; only one strain (GBS1428) was capsular type III/ST-17. The fbsB and hylB genes were detected in all isolates, while the iag, lmb, and fbsA genes were detected in 94.5%, 91%, and 91% of oncological isolates, respectively. The combination of PI-1 and PI-2a was the most common (60%) among S. agalactiae strains isolated from oncologic patients. S. agalactiae strains were resistant to tetracycline (85.5%), erythromycin (9%), and clindamycin (5.5%). Norfloxacin non-susceptible was detected in 7.3% of S. agalactiae strains. Our findings reinforce the need for S. agalactiae control measures in Brazil, including cancer patients.


Assuntos
Neoplasias/complicações , Infecções Estreptocócicas/epidemiologia , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/genética , Streptococcus agalactiae/isolamento & purificação , Adolescente , Adulto , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Brasil/epidemiologia , Farmacorresistência Bacteriana Múltipla , Genótipo , Humanos , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Neoplasias/microbiologia , Streptococcus agalactiae/efeitos dos fármacos , Streptococcus agalactiae/patogenicidade , Fatores de Virulência/genética , Adulto Jovem
7.
Microb Drug Resist ; 26(12): 1546-1558, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32429830

RESUMO

Disinfection and antisepsis are of primary importance in controlling nosocomial infections and outbreaks by pathogens expressing multiple resistance to antimicrobial agents (multidrug-resistant [MDR]) used in therapy. Nowadays, infections related to health services (HAIs) due to MDR and multidrug-susceptible (MDS) Corynebacterium striatum should not be underestimated, including patients using invasive medical devices. The virulence potential of C. striatum needs further investigation. Currently, susceptibility profiles of planktonic and/or sessile forms of four C. striatum strains of different pulsed-field gel electrophoresis types were examined as biocides based on the manufacturer's recommendations: 2% glutaraldehyde (GA), 2% peracetic acid (PA), 1% potassium monopersulfate (Virkon®; VK), 1% sodium hypochlorite (SH), and 70% ethyl alcohol (ET). Time-kill assays using 2% bovine serum albumin (BSA) were performed for evaluation of influence of organic matter on biocides effects. Planktonic forms expressed GA resistance at different levels. C. striatum viability was observed until 2, 4, 20, and 30 min for MDR 2369/II, MDS 1954/IV, MDR 1987/I, and MDS 1961/III strains, respectively. In contrast to GA, the biocides PA, VK24h, SH, and ET had higher effective bacterial mortality. However, storage of VK (48 hr) reduced their biocide activities. Moreover, mature biofilms were produced on abiotic substrates, including steel surfaces. Post-treatment with GA (30 min), survival of sessile forms was ≥100% than planktonic forms of all C. striatum tested strains. Independent of biocides tested, BSA increased the survival of planktonic and sessile forms (p ≤ 0.005). Present data indicated that hospital staff should be aware of dissemination and eradication of HAIs by C. striatum presenting resistance to biocides, including high-level disinfectants, such as GA.


Assuntos
Anti-Infecciosos Locais/farmacologia , Biofilmes/efeitos dos fármacos , Corynebacterium/efeitos dos fármacos , Desinfetantes/farmacologia , Farmacorresistência Bacteriana Múltipla , Plâncton/efeitos dos fármacos , Adulto , Infecção Hospitalar/prevenção & controle , Eletroforese em Gel de Campo Pulsado , Feminino , Humanos , Masculino , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Virulência
8.
Mem Inst Oswaldo Cruz ; 115: e190398, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32187326

RESUMO

BACKGROUND: Streptococcus agalactiae capsular type III strains are a leading cause of invasive neonatal infections. Many pathogens have developed mechanisms to escape from host defense response using the host membrane microdomain machinery. Lipid rafts play an important role in a variety of cellular functions and the benefit provided by interaction with lipid rafts can vary from one pathogen to another. OBJECTIVES: This study aims to evaluate the involvement of membrane microdomains during infection of human endothelial cell by S. agalactiae. METHODS: The effects of cholesterol depletion and PI3K/AKT signaling pathway activation during S. agalactiae-human umbilical vein endothelial cells (HUVEC) interaction were analysed by pre-treatment with methyl-ß-cyclodextrin (MßCD) or LY294002 inhibitors, immunofluorescence and immunoblot analysis. The involvement of lipid rafts was analysed by colocalisation of bacteria with flotillin-1 and caveolin-1 using fluorescence confocal microscopy. FINDINGS: In this work, we demonstrated the importance of the integrity of lipid rafts microdomains and activation of PI3K/Akt pathway during invasion of S. agalactiae strain to HUVEC cells. Our results suggest the involvement of flotillin-1 and caveolin-1 during the invasion of S. agalactiae strain in HUVEC cells. CONCLUSIONS: The collection of our results suggests that lipid microdomain affects the interaction of S. agalactiae type III belonging to the hypervirulent ST-17 with HUVEC cells through PI3K/Akt signaling pathway.


Assuntos
Células Endoteliais/virologia , Lipídeos de Membrana , Microdomínios da Membrana/virologia , Streptococcus agalactiae/patogenicidade , Virulência , Humanos , Recém-Nascido , Streptococcus agalactiae/genética
9.
Infect Genet Evol ; 80: 104195, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31954181

RESUMO

Streptococcus agalactiae are important pathogenic bacteria that cause severe infections in humans, especially neonates. The mechanism by which ST-17 causes invasive infections than other STs is not well understood. In this study, we sequenced the first genome of a S. agalactiae ST-17 strain isolated in Brazil using the Illumina HiSeq 2500 technology. S. agalactiae GBS90356 ST-17 belongs to the capsular type III and was isolated from a neonatal with a fatal case of meningitis. The genome presented a size of 2.03 Mbp and a G + C content of 35.2%. S. agalactiae has 706 genes in its core genome and an open pan-genome with a size of 5.020 genes, suggesting a high genomic plasticity. GIPSy software was used to identify 10 Pathogenicity islands (PAIs) which corresponded to 15% of the genome size. IslandViewer4 corroborated the prediction of six PAIs. The pathogenicity islands showed important virulence factors genes for S. agalactiae e.g. neu, cps, dlt, fbs, cfb, lmb. SignalP detected 20 proteins with signal peptides among the 352 proteins found in PAIs, which 60% were located in the SagPAI_5. SagPAI_2 and 5 were mainly detected in ST-17 strains studied. Moreover, we identified 51 unique genes, 9 recombination regions and a large number of SNPs with an average of 760.3 polymorphisms, which can be related with high genomic plasticity and virulence during host-pathogen interactions. Our results showed implications for pathogenesis, evolution, concept of species and in silico analysis value to understand the epidemiology and genome plasticity of S. agalactiae.


Assuntos
Genoma Bacteriano , Genômica , Infecções Estreptocócicas/epidemiologia , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/classificação , Streptococcus agalactiae/genética , Brasil/epidemiologia , Biologia Computacional/métodos , Genômica/métodos , Humanos , Anotação de Sequência Molecular , Filogenia , Vigilância em Saúde Pública , Streptococcus agalactiae/isolamento & purificação , Streptococcus agalactiae/patogenicidade , Virulência/genética , Fatores de Virulência/genética
10.
Mem Inst Oswaldo Cruz ; 113(6): e140421, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29641644

RESUMO

BACKGROUND Streptococcus agalactiae can causes sepsis, pneumonia, and meningitis in neonates, the elderly, and immunocompromised patients. Although the virulence properties of S. agalactiae have been partially elucidated, the molecular mechanisms related to reactive oxygen species (ROS) generation in infected human endothelial cells need further investigation. OBJECTIVES This study aimed to evaluate the influence of oxidative stress in human umbilical vein endothelial cells (HUVECs) during S. agalactiae infection. METHODS ROS production during S. agalactiae-HUVEC infection was detected using the probe CM-H2DCFDA. Microfilaments labelled with phalloidin-FITC and p47phox-Alexa 546 conjugated were analysed by immunofluorescence. mRNA levels of p47phox (NADPH oxidase subunit) were assessed using Real Time qRT-PCR. The adherence and intracellular viability of S. agalactiae in HUVECs with or without pre-treatment of DPI, apocynin (NADPH oxidase inhibitors), and LY294002 (PI3K inhibitor) were evaluated by penicillin/gentamicin exclusion. Phosphorylation of p47phox and Akt activation by S. agalactiae were evaluated by immunoblotting analysis. FINDINGS Data showed increased ROS production 15 min after HUVEC infection. Real-Time qRT-PCR and western blotting performed in HUVEC infected with S. agalactiae detected alterations in mRNA levels and activation of p47phox. Pre-treatment of endothelial cells with NADPH oxidase (DPI and apocynin) and PI3K/Akt pathway (LY294002) inhibitors reduced ROS production, bacterial intracellular viability, and generation of actin stress fibres in HUVECs infected with S. agalactiae. CONCLUSIONS ROS generation via the NADPH oxidase pathway contributes to invasion of S. agalactiae in human endothelial cells accompanied by cytoskeletal reorganisation through the PI3K/Akt pathway, which provides novel evidence for the involvement of oxidative stress in S. agalactiae pathogenesis.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , NADPH Oxidases/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Infecções Estreptocócicas/metabolismo , Streptococcus agalactiae/metabolismo , Análise de Variância , Aderência Bacteriana , Western Blotting , Células Endoteliais da Veia Umbilical Humana/microbiologia , Humanos , NADPH Oxidases/análise , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/análise , Reação em Cadeia da Polimerase em Tempo Real , Valores de Referência , Transdução de Sinais/fisiologia , Fatores de Tempo
11.
Mem. Inst. Oswaldo Cruz ; 113(6): e140421, 2018. graf
Artigo em Inglês | LILACS | ID: biblio-894933

RESUMO

BACKGROUND Streptococcus agalactiae can causes sepsis, pneumonia, and meningitis in neonates, the elderly, and immunocompromised patients. Although the virulence properties of S. agalactiae have been partially elucidated, the molecular mechanisms related to reactive oxygen species (ROS) generation in infected human endothelial cells need further investigation. OBJECTIVES This study aimed to evaluate the influence of oxidative stress in human umbilical vein endothelial cells (HUVECs) during S. agalactiae infection. METHODS ROS production during S. agalactiae-HUVEC infection was detected using the probe CM-H2DCFDA. Microfilaments labelled with phalloidin-FITC and p47phox-Alexa 546 conjugated were analysed by immunofluorescence. mRNA levels of p47phox (NADPH oxidase subunit) were assessed using Real Time qRT-PCR. The adherence and intracellular viability of S. agalactiae in HUVECs with or without pre-treatment of DPI, apocynin (NADPH oxidase inhibitors), and LY294002 (PI3K inhibitor) were evaluated by penicillin/gentamicin exclusion. Phosphorylation of p47phox and Akt activation by S. agalactiae were evaluated by immunoblotting analysis. FINDINGS Data showed increased ROS production 15 min after HUVEC infection. Real-Time qRT-PCR and western blotting performed in HUVEC infected with S. agalactiae detected alterations in mRNA levels and activation of p47phox. Pre-treatment of endothelial cells with NADPH oxidase (DPI and apocynin) and PI3K/Akt pathway (LY294002) inhibitors reduced ROS production, bacterial intracellular viability, and generation of actin stress fibres in HUVECs infected with S. agalactiae. CONCLUSIONS ROS generation via the NADPH oxidase pathway contributes to invasion of S. agalactiae in human endothelial cells accompanied by cytoskeletal reorganisation through the PI3K/Akt pathway, which provides novel evidence for the involvement of oxidative stress in S. agalactiae pathogenesis.


Assuntos
Humanos , Espécies Reativas de Oxigênio/análise , NADPH Oxidases/análise , NADPH Oxidases/metabolismo , Células Endoteliais da Veia Umbilical Humana/microbiologia , Transdução de Sinais/fisiologia , Reação em Cadeia da Polimerase em Tempo Real
12.
Stand Genomic Sci ; 11: 39, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27274785

RESUMO

Streptococcus agalactiae, also referred to as Group B Streptococcus, is a frequent resident of the rectovaginal tract in humans, and a major cause of neonatal infection. The pathogen can also infect adults with underlying disease, particularly the elderly and immunocompromised ones. In addition, S. agalactiae is a known fish pathogen, which compromises food safety and represents a zoonotic hazard. This study provides valuable structural, functional and evolutionary genomic information of a human S. agalactiae serotype Ia (ST-103) GBS85147 strain isolated from the oropharynx of an adult patient from Rio de Janeiro, thereby representing the first human isolate in Brazil. We used the Ion Torrent PGM platform with the 200 bp fragment library sequencing kit. The sequencing generated 578,082,183 bp, distributed among 2,973,022 reads, resulting in an approximately 246-fold mean coverage depth and was assembled using the Mira Assembler v3.9.18. The S. agalactiae strain GBS85147 comprises of a circular chromosome with a final genome length of 1,996,151 bp containing 1,915 protein-coding genes, 18 rRNA, 63 tRNA, 2 pseudogenes and a G + C content of 35.48 %.

13.
Antonie Van Leeuwenhoek ; 107(6): 1387-99, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25828766

RESUMO

Biofilm-related infections are considered a major cause of morbidity and mortality in hospital environments. Biofilms allow microorganisms to exchange genetic material and to become persistent colonizers and/or multiresistant to antibiotics. Corynebacterium pseudodiphtheriticum (CPS), a commensal bacterium that colonizes skin and mucosal sites has become progressively multiresistant and responsible for severe nosocomial infections. However, virulence factors of this emergent pathogen remain unclear. Herein, we report the adhesive properties and biofilm formation on hydrophilic (glass) and hydrophobic (plastic) abiotic surfaces by CPS strains isolated from patients with localized (ATCC10700/Pharyngitis) and systemic (HHC1507/Bacteremia) infections. Adherence to polystyrene attributed to hydrophobic interactions between bacterial cells and this negatively charged surface indicated the involvement of cell surface hydrophobicity in the initial stage of biofilm formation. Attached microorganisms multiplied and formed microcolonies that accumulated as multilayered cell clusters, a step that involved intercellular adhesion and synthesis of extracellular matrix molecules. Further growth led to the formation of dense bacterial aggregates embedded in the exopolymeric matrix surrounded by voids, typical of mature biofilms. Data also showed CPS recognizing human fibrinogen (Fbg) and fibronectin (Fn) and involvement of these sera components in formation of "conditioning films". These findings suggested that biofilm formation may be associated with the expression of different adhesins. CPS may form biofilms in vivo possibly by an adherent biofilm mode of growth in vitro currently demonstrated on hydrophilic and hydrophobic abiotic surfaces. The affinity to Fbg and Fn and the biofilm-forming ability may contribute to the establishment and dissemination of infection caused by CPS.


Assuntos
Aderência Bacteriana , Biofilmes/crescimento & desenvolvimento , Corynebacterium/fisiologia , Microbiologia Ambiental , Fibrinogênio/metabolismo , Fibronectinas/metabolismo , Adesinas Bacterianas/metabolismo , Bacteriemia/microbiologia , Técnicas Bacteriológicas , Corynebacterium/crescimento & desenvolvimento , Corynebacterium/isolamento & purificação , Infecções por Corynebacterium/microbiologia , Vidro , Humanos , Microscopia , Faringite/microbiologia , Plásticos
14.
Mem Inst Oswaldo Cruz ; 109(7): 940-3, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25410999

RESUMO

Endothelial dysfunction is a major component of the pathophysiology of septicaemic group B Streptococcus (GBS) infections. Although cytokines have been shown to activate human umbilical vein endothelial cells (HUVECs), the capacity of interferon (IFN)-γ to enhance the microbicidal activity of HUVECs against GBS has not been studied. We report that the viability of intracellular bacteria was reduced in HUVECs activated by IFN-γ. Enhanced fusion of lysosomes with bacteria-containing vacuoles was observed by acid phosphatase and the colocalisation of Rab-5, Rab-7 and lysosomal-associated membrane protein-1 with GBS in IFN-γ-activated HUVECs. IFN-γ resulted in an enhancement of the phagosome maturation process in HUVECs, improving the capacity to control the intracellular survival of GBS.


Assuntos
Anti-Infecciosos/farmacologia , Células Endoteliais da Veia Umbilical Humana/microbiologia , Interferon gama/farmacologia , Viabilidade Microbiana/efeitos dos fármacos , Infecções Estreptocócicas/tratamento farmacológico , Streptococcus agalactiae/efeitos dos fármacos , Fosfatase Ácida/metabolismo , Aderência Bacteriana/efeitos dos fármacos , Endocitose , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Lisossomos/efeitos dos fármacos , Fagossomos/efeitos dos fármacos , Cultura Primária de Células , Infecções Estreptocócicas/prevenção & controle , Análise de Sobrevida
15.
Mem. Inst. Oswaldo Cruz ; 109(7): 940-943, 11/2014. graf
Artigo em Inglês | LILACS | ID: lil-728811

RESUMO

Endothelial dysfunction is a major component of the pathophysiology of septicaemic group B Streptococcus (GBS) infections. Although cytokines have been shown to activate human umbilical vein endothelial cells (HUVECs), the capacity of interferon (IFN)-γ to enhance the microbicidal activity of HUVECs against GBS has not been studied. We report that the viability of intracellular bacteria was reduced in HUVECs activated by IFN-γ. Enhanced fusion of lysosomes with bacteria-containing vacuoles was observed by acid phosphatase and the colocalisation of Rab-5, Rab-7 and lysosomal-associated membrane protein-1 with GBS in IFN-γ-activated HUVECs. IFN-γ resulted in an enhancement of the phagosome maturation process in HUVECs, improving the capacity to control the intracellular survival of GBS.


Assuntos
Humanos , Anti-Infecciosos/farmacologia , Células Endoteliais da Veia Umbilical Humana/microbiologia , Interferon gama/farmacologia , Viabilidade Microbiana/efeitos dos fármacos , Infecções Estreptocócicas/tratamento farmacológico , Streptococcus agalactiae/efeitos dos fármacos , Fosfatase Ácida/metabolismo , Aderência Bacteriana/efeitos dos fármacos , Endocitose , Células Endoteliais da Veia Umbilical Humana/metabolismo , Lisossomos/efeitos dos fármacos , Cultura Primária de Células , Fagossomos/efeitos dos fármacos , Análise de Sobrevida , Infecções Estreptocócicas/prevenção & controle
16.
Microbiology (Reading) ; 160(Pt 3): 537-546, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24344208

RESUMO

Although infection by Corynebacterium diphtheriae is a model of extracellular mucosal pathogenesis, different clones have been also associated with invasive infections such as sepsis, endocarditis, septic arthritis and osteomyelitis. The mechanisms that promote C. diphtheriae infection and haematogenic dissemination need further investigation. In this study we evaluated the association and invasion mechanisms with human umbilical vein endothelial cells (HUVECs) and experimental arthritis in mice of endocarditis-associated strains and control non-invasive strains. C. diphtheriae strains were able to adhere to and invade HUVECs at different levels. The endocarditis-associated strains displayed an aggregative adherence pattern and a higher number of internalized viable cells in HUVECs. Transmission electron microscopy (TEM) analysis revealed intracellular bacteria free in the cytoplasm and/or contained in a host-membrane-confined compartment as single micro-organisms. Data showed bacterial internalization dependent on microfilament and microtubule stability and involvement of protein phosphorylation in the HUVEC signalling pathway. A high number of affected joints and high arthritis index in addition to the histopathological features indicated a strain-dependent ability of C. diphtheriae to cause severe polyarthritis. A correlation between the arthritis index and increased systemic levels of IL-6 and TNF-α was observed for endocarditis-associated strains. In conclusion, higher incidence of potential mechanisms by which C. diphtheriae may access the bloodstream through the endothelial barrier and stimulate the production of pro-inflammatory cytokines such as IL-6 and TNF-α, in addition to the ability to affect the joints and induce arthritis through haematogenic spread are thought to be related to the pathogenesis of endocarditis-associated strains.


Assuntos
Corynebacterium diphtheriae/fisiologia , Endocardite/microbiologia , Células Endoteliais/microbiologia , Animais , Artrite/microbiologia , Aderência Bacteriana , Linhagem Celular , Citocinas/biossíntese , Endocardite/metabolismo , Células Endoteliais/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/microbiologia , Humanos , Camundongos
17.
Antonie Van Leeuwenhoek ; 104(6): 1125-33, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24052367

RESUMO

We explored Group B Streptococcus (GBS)-induced apoptosis in human umbilical vein endothelial cells (HUVEC) and the role of phosphoramidon, a zinc metalloprotease inhibitor, in this process. GBS 90186 strain (serotype V, a blood isolate) and concentrated supernatant (CS) were used to investigate the viability and morphological alterations in HUVEC by Trypan blue uptake, electrophoresis in 2 % agarose gel and scanning electron microscopy assays. Apoptosis before and after phosphoramidon-treatment were verified by flow cytometry using annexin V-FITC labeling. Differences were considered significant when P < 0.05 using unpaired Student's t test. GBS and CS induced HUVEC death by apoptosis (76.5 and 32 %, respectively) with an increasing pro-apoptotic Bax expression and decreasing anti-apoptotic Bcl-2 expression. Caspase-3 was activated during GBS-induced endothelial apoptosis. Phosphoramidon reduced 89.3 and 100 % of GBS and CS cell death by apoptosis, respectively. Some GBS strains may induce cell death by apoptosis with involvement of metalloproteases and signaling through the intrinsic pathway of apoptosis, which may contribute to GBS survival during sepsis of adults and neonates.


Assuntos
Apoptose , Células Endoteliais/microbiologia , Células Endoteliais/fisiologia , Glicopeptídeos/metabolismo , Metaloproteases/metabolismo , Inibidores de Proteases/metabolismo , Streptococcus agalactiae/enzimologia , Anexina A5/análise , Sobrevivência Celular , Células Cultivadas , Eletroforese , Humanos , Metaloproteases/antagonistas & inibidores , Microscopia Eletrônica , Coloração e Rotulagem/métodos , Azul Tripano/metabolismo
18.
Braz. j. microbiol ; 44(3): 869-872, July-Sept. 2013. tab
Artigo em Inglês | LILACS | ID: lil-699819

RESUMO

GBS serotypes III and V were the most prevalent in pregnant women and exhibited resistance to tetracycline, clindamycin and sulfamethoxazole/trimethoprim. Serotype III showed high sialic acid content and PFGE analysis discerned 33 heterogeneous profiles. Phenotypic and genotypic characterization could be relevant to control GBS infections unaffected by intra-partum chemoprophylaxis.


Assuntos
Feminino , Humanos , Recém-Nascido , Gravidez , Complicações Infecciosas na Gravidez/epidemiologia , Sorogrupo , Infecções Estreptocócicas/epidemiologia , Streptococcus agalactiae/isolamento & purificação , Antibacterianos/farmacologia , Brasil/epidemiologia , Análise por Conglomerados , Farmacorresistência Bacteriana , Eletroforese em Gel de Campo Pulsado , Genótipo , Tipagem Molecular , Prevalência , Complicações Infecciosas na Gravidez/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/classificação , Streptococcus agalactiae/efeitos dos fármacos
19.
20.
Mem Inst Oswaldo Cruz ; 107(4): 486-93, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22666859

RESUMO

Corynebacterium pseudodiphtheriticum is a well-known human pathogen that mainly causes respiratory disease and is associated with high mortality in compromised hosts. Little is known about the virulence factors and pathogenesis of C. pseudodiphtheriticum. In this study, cultured human epithelial (HEp-2) cells were used to analyse the adherence pattern, internalisation and intracellular survival of the ATCC 10700 type strain and two additional clinical isolates. These microorganisms exhibited an aggregative adherence-like pattern to HEp-2 cells characterised by clumps of bacteria with a "stacked-brick" appearance. The differences in the ability of these microorganisms to invade and survive within HEp-2 cells and replicate in the extracellular environment up to 24 h post infection were evaluated. The fluorescent actin staining test demonstrated that actin polymerisation is involved in the internalisation of the C. pseudodiphtheriticum strains. The depolymerisation of microfilaments by cytochalasin E significantly reduced the internalisation of C. pseudodiphtheriticum by HEp-2 cells. Bacterial internalisation and cytoskeletal rearrangement seemed to be partially triggered by the activation of tyrosine kinase activity. Although C. pseudodiphtheriticum strains did not demonstrate an ability to replicate intracellularly, HEp-2 cells were unable to fully clear the pathogen within 24 h. These characteristics may explain how some C. pseudodiphtheriticum strains cause severe infection in human patients.


Assuntos
Aderência Bacteriana/fisiologia , Corynebacterium/patogenicidade , Células Epiteliais/microbiologia , Corynebacterium/fisiologia , Células Hep G2 , Humanos , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...