Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Int J Mol Sci ; 25(4)2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38396905

RESUMO

Chronic rhinosinusitis (CRS) is a multifactorial infection of the nasal cavity and sinuses. In this study, nasal swabs from control donors (N = 128) and patients with CRS (N = 246) were analysed. Culture methods and metagenomics revealed no obvious differences in the composition of the bacterial communities between the two groups. However, at the functional level, several metabolic pathways were significantly enriched in the CRS group compared to the control group. Pathways such as carbohydrate transport metabolism, ATP synthesis, cofactors and vitamins, photosynthesis and transcription were highly enriched in CRS. In contrast, pathways related to lipid metabolism were more representative in the control microbiome. As S. aureus is one of the main species found in the nasal cavity, staphylococcal isolates from control and CRS samples were analysed by microarray and functional assays. Although no significant genetic differences were detected by microarray, S. aureus from CRS induced less cytotoxicity to lung cells and lower rates of glycolysis in host cells than control isolates. These results suggest the differential modulation of staphylococcal virulence by the environment created by other microorganisms and their interactions with host cells in control and CRS samples. These changes were reflected in the differential expression of cytokines and in the expression of Agr, the most important quorum-sensing regulator of virulence in S. aureus. In addition, the CRS isolates remained stable in their cytotoxicity, whereas the cytotoxic activity of S. aureus isolated from control subjects decreased over time during in vitro passage. These results suggest that host factors influence the virulence of S. aureus and promote its adaptation to the nasal environment during CRS.


Assuntos
Seios Paranasais , Rinite , Rinossinusite , Sinusite , Infecções Estafilocócicas , Humanos , Staphylococcus aureus/genética , Adaptação ao Hospedeiro , Sinusite/microbiologia , Infecções Estafilocócicas/microbiologia , Doença Crônica , Rinite/microbiologia
2.
Int J Mol Sci ; 24(22)2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-38003500

RESUMO

Methicillin-sensitive Staphylococcus (S.) aureus (MSSA) bacteremia remains a global challenge, despite the availability of antibiotics. Primary treatments include ß-lactam agents such as cefazolin and flucloxacillin. Ongoing discussions have focused on the potential synergistic effects of combining these agents with rifampicin or fosfomycin to combat infections associated with biofilm formation. Managing staphylococcal infections is challenging due to antibacterial resistance, biofilms, and S. aureus's ability to invade and replicate within host cells. Intracellular invasion shields the bacteria from antibacterial agents and the immune system, often leading to incomplete bacterial clearance and chronic infections. Additionally, S. aureus can assume a dormant phenotype, known as the small colony variant (SCV), further complicating eradication and promoting persistence. This study investigated the impact of antibiotic combinations on the persistence of S. aureus 6850 and its stable small colony variant (SCV strain JB1) focusing on intracellular survival and biofilm formation. The results from the wild-type strain 6850 demonstrate that ß-lactams combined with RIF effectively eliminated biofilms and intracellular bacteria but tend to select for SCVs in planktonic culture and host cells. Higher antibiotic concentrations were associated with an increase in the zeta potential of S. aureus, suggesting reduced membrane permeability to antimicrobials. When using the stable SCV mutant strain JB1, antibiotic combinations with rifampicin successfully cleared planktonic bacteria and biofilms but failed to eradicate intracellular bacteria. Given these findings, it is reasonable to report that ß-lactams combined with rifampicin represent the optimal treatment for MSSA bacteremia. However, caution is warranted when employing this treatment over an extended period, as it may elevate the risk of selecting for small colony variants (SCVs) and, consequently, promoting bacterial persistence.


Assuntos
Bacteriemia , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Humanos , Antibacterianos/farmacologia , Staphylococcus aureus , Meticilina/farmacologia , Rifampina/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Biofilmes , beta-Lactamas/farmacologia , Testes de Sensibilidade Microbiana
3.
Int J Mol Sci ; 24(11)2023 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-37298718

RESUMO

Osteomyelitis is an infection of the bone that is often difficult to treat and causes a significant healthcare burden. Staphylococcus aureus is the most common pathogen causing osteomyelitis. Osteomyelitis mouse models have been established to gain further insights into the pathogenesis and host response. Here, we use an established S. aureus hematogenous osteomyelitis mouse model to investigate morphological tissue changes and bacterial localization in chronic osteomyelitis with a focus on the pelvis. X-ray imaging was performed to follow the disease progression. Six weeks post infection, when osteomyelitis had manifested itself with a macroscopically visible bone deformation in the pelvis, we used two orthogonal methods, namely fluorescence imaging and label-free Raman spectroscopy, to characterise tissue changes on a microscopic scale and to localise bacteria in different tissue regions. Hematoxylin and eosin as well as Gram staining were performed as a reference method. We could detect all signs of a chronically florid tissue infection with osseous and soft tissue changes as well as with different inflammatory infiltrate patterns. Large lesions dominated in the investigated tissue samples. Bacteria were found to form abscesses and were distributed in high numbers in the lesion, where they could occasionally also be detected intracellularly. In addition, bacteria were found in lower numbers in surrounding muscle tissue and even in lower numbers in trabecular bone tissue. The Raman spectroscopic imaging revealed a metabolic state of the bacteria with reduced activity in agreement with small cell variants found in other studies. In conclusion, we present novel optical methods to characterise bone infections, including inflammatory host tissue reactions and bacterial adaptation.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Osteomielite , Infecções Estafilocócicas , Camundongos , Animais , Staphylococcus aureus/fisiologia , Osteomielite/patologia , Modelos Animais de Doenças , Inflamação , Infecções Estafilocócicas/microbiologia , Infecção Persistente
4.
Immunology ; 166(1): 47-67, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35143048

RESUMO

Staphylococcus aureus causes severe infections associated with inflammation, such as sepsis or osteomyelitis. Inflammatory processes are regulated by distinct lipid mediators (LMs) but how their biosynthetic pathways are orchestrated in S. aureus infections is elusive. We show that S. aureus strikingly not only modulates pro-inflammatory, but also inflammation-resolving LM pathways in murine osteomyelitis and osteoclasts as well as in human monocyte-derived macrophages (MDMs) with different phenotype. Targeted LM metabololipidomics using ultra-performance liquid chromatography-tandem mass spectrometry revealed massive generation of LM with distinct LM signature profiles in acute and chronic phases of S. aureus-induced murine osteomyelitis in vivo. In human MDM, S. aureus elevated cyclooxygenase-2 (COX-2) and microsomal prostaglandin E2  synthase-1 (mPGES-1), but impaired the levels of 15-lipoxygenase-1 (15-LOX-1), with respective changes in LM signature profiles initiated by these enzymes, that is, elevated PGE2 and impaired specialized pro-resolving mediators, along with reduced M2-like phenotypic macrophage markers. The cell wall component, lipoteichoic acid (LTA), mimicked the impact of S. aureus elevating COX-2/mPGES-1 expression via NF-κB and p38 MAPK signalling in MDM, while the impairment of 15-LOX-1 correlates with reduced expression of Lamtor1. In conclusion, S. aureus dictates LM pathways via LTA resulting in a shift from anti-inflammatory M2-like towards pro-inflammatory M1-like LM signature profiles.


Assuntos
Osteomielite , Staphylococcus aureus , Animais , Ciclo-Oxigenase 2/metabolismo , Dinoprostona , Inflamação/metabolismo , Lipopolissacarídeos , Camundongos , Prostaglandina-E Sintases/metabolismo , Receptores Depuradores Classe E , Ácidos Teicoicos
5.
Cell Mol Life Sci ; 77(19): 3841-3858, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31807813

RESUMO

Massive neutrophil infiltration is an early key event in infectious inflammation, accompanied by chemotactic leukotriene (LT)B4 generation. LTB4 biosynthesis is mediated by 5-lipoxygenase (5-LOX), but which pathogenic factors cause 5-LOX activation during bacterial infections is elusive. Here, we reveal staphylococcal exotoxins as 5-LOX activators. Conditioned medium of wild-type Staphylococcus aureus but not of exotoxin-deficient strains induced 5-LOX activation in transfected HEK293 cells. Two different staphylococcal exotoxins mimicked the effects of S. aureus-conditioned medium: (1) the pore-forming toxin α-hemolysin and (2) amphipathic α-helical phenol-soluble modulin (PSM) peptides. Interestingly, in human neutrophils, 5-LOX activation was exclusively evoked by PSMs, which was prevented by the selective FPR2/ALX receptor antagonist WRW4. 5-LOX activation by PSMs was confirmed in vivo as LT formation in infected paws of mice was impaired in response to PSM-deficient S. aureus. Conclusively, exotoxins from S. aureus are potent pathogenic factors that activate 5-LOX and induce LT formation in neutrophils.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Ativação Enzimática/efeitos dos fármacos , Exotoxinas/farmacologia , Leucotrienos/biossíntese , Staphylococcus aureus/metabolismo , Animais , Toxinas Bacterianas/farmacologia , Cálcio/metabolismo , Doenças do Pé/metabolismo , Doenças do Pé/patologia , Doenças do Pé/veterinária , Células HEK293 , Proteínas Hemolisinas/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Oligopeptídeos/farmacologia , Receptores de Lipoxinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/patologia , Infecções Estafilocócicas/veterinária , Staphylococcus aureus/patogenicidade
6.
Curr Microbiol ; 76(7): 842-847, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31053906

RESUMO

Diabetic foot ulcer infections are frequently polymicrobial in nature and exhibit increased morbidity and mortality, as well as, treatment failures. Interactions between Acinetobacter baumannii and Staphylococcus aureus were studied, which showed strain-dependent changes in growth and antibiotic susceptibility. This study examined the interactions between two clinical strains of A. baumannii (1929) and S. aureus (1928) that were recovered from skin and soft tissues of a diabetic patient. When S. aureus 1928 and A. baumannii 1929 were co-cultured together, there was no significant decrease in growth in either clinical strains, indicating that both strains can co-exist in the same site of infection. Additionally, neither strains experienced statistically significant changes in susceptibility. These findings highlight that these two pathogens can be found in the same niche of infection, which may lead to more aggressive outcome of the infection.


Assuntos
Infecções por Acinetobacter/microbiologia , Coinfecção/microbiologia , Pé Diabético/microbiologia , Interações Microbianas , Infecções dos Tecidos Moles/microbiologia , Infecções Estafilocócicas/microbiologia , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/crescimento & desenvolvimento , Acinetobacter baumannii/isolamento & purificação , Acinetobacter baumannii/fisiologia , Antibacterianos/farmacologia , Complicações do Diabetes/microbiologia , Humanos , Testes de Sensibilidade Microbiana , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/isolamento & purificação , Staphylococcus aureus/fisiologia
7.
Int J Med Microbiol ; 308(7): 761-769, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29843979

RESUMO

Staphylococcus aureus is the most frequent pathogen causing diabetic foot infections. Here, we investigated the degree of bacterial virulence required to establish invasive tissue infections in diabetic organisms. Staphylococcal isolates from diabetic and non-diabetic foot ulcers were tested for their virulence in in vitro functional assays of host cell invasion and cytotoxicity. Isolates from diabetes mellitus type I/II patients exhibited less virulence than isolates from non-diabetic patients, but were nevertheless able to establish severe infections. In some cases, non-invasive isolates were detected deep within diabetic wounds, even though the strains were non-pathogenic in cell culture models. Testing of defined isolates in murine footpad injection models revealed that both low- and high-virulent bacterial strains persisted in higher numbers in diabetic compared to non-diabetic hosts, suggesting that hyperglycemia favors bacterial survival. Additionally, the bacterial load was higher in NOD mice, which have a compromised immune system, compared to C57Bl/6 mice. Our results reveal that high as well as low-virulent staphylococcal strains are able to cause soft tissue infections and to persist in diabetic humans and mice, suggesting a reason for the frequent and endangering infections in patients with diabetes.


Assuntos
Carga Bacteriana , Diabetes Mellitus Experimental/patologia , Pé Diabético/microbiologia , Infecções dos Tecidos Moles/microbiologia , Staphylococcus aureus/patogenicidade , Adulto , Idoso , Animais , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana , Humanos , Hiperglicemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Pessoa de Meia-Idade , Staphylococcus aureus/isolamento & purificação , Estreptozocina , Virulência
8.
Am J Pathol ; 187(2): 268-279, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28088288

RESUMO

Staphylococcus aureus causes very serious infections of vascular grafts. Knowledge of the molecular mechanisms of this disease is largely lacking because of the absence of representable models. Therefore, the aim of this study was to set up a mouse model of vascular graft infections that closely mimics the human situation. A catheter was inserted into the right carotid artery of mice, which acted as a vascular graft. Mice were infected i.v. using 8 different S. aureus strains, and development of the infection was followed up. Although all strains had varying abilities to form biofilm in vitro and different levels of virulence in mice, they all caused biofilm formation on the grafts. This graft infection was monitored using magnetic resonance imaging (MRI) and 18F-fluordeoxyglucose positron emission tomography (FDG-PET). MRI allowed the quantification of blood flow through the arteries, which was decreased in the catheter after infection. FDG-PET revealed high inflammation levels at the site of the catheter after infection. This model closely resembles the situation in patients, which is characterized by a tight interplay between pathogen and host, and can therefore be used for the testing of novel treatment, diagnosis, and prevention strategies. In addition, combining MRI and PET with microscopic techniques provides an appropriate way to characterize the course of these infections and to precisely analyze biofilm development.


Assuntos
Biofilmes/crescimento & desenvolvimento , Infecções Relacionadas a Cateter/diagnóstico por imagem , Modelos Animais de Doenças , Infecções Estafilocócicas/diagnóstico por imagem , Animais , Prótese Vascular/microbiologia , Ensaio de Imunoadsorção Enzimática , Hibridização in Situ Fluorescente , Imageamento por Ressonância Magnética , Camundongos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Tomografia por Emissão de Pósitrons , Staphylococcus aureus
9.
PLoS Pathog ; 11(4): e1004870, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25923704

RESUMO

Staphylococcus aureus is a major human pathogen that causes a range of infections from acute invasive to chronic and difficult-to-treat. Infection strategies associated with persisting S. aureus infections are bacterial host cell invasion and the bacterial ability to dynamically change phenotypes from the aggressive wild-type to small colony variants (SCVs), which are adapted for intracellular long-term persistence. The underlying mechanisms of the bacterial switching and adaptation mechanisms appear to be very dynamic, but are largely unknown. Here, we analyzed the role and the crosstalk of the global S. aureus regulators agr, sarA and SigB by generating single, double and triple mutants, and testing them with proteome analysis and in different in vitro and in vivo infection models. We were able to demonstrate that SigB is the crucial factor for adaptation in chronic infections. During acute infection, the bacteria require the simultaneous action of the agr and sarA loci to defend against invading immune cells by causing inflammation and cytotoxicity and to escape from phagosomes in their host cells that enable them to settle an infection at high bacterial density. To persist intracellularly the bacteria subsequently need to silence agr and sarA. Indeed agr and sarA deletion mutants expressed a much lower number of virulence factors and could persist at high numbers intracellularly. SigB plays a crucial function to promote bacterial intracellular persistence. In fact, ΔsigB-mutants did not generate SCVs and were completely cleared by the host cells within a few days. In this study we identified SigB as an essential factor that enables the bacteria to switch from the highly aggressive phenotype that settles an acute infection to a silent SCV-phenotype that allows for long-term intracellular persistence. Consequently, the SigB-operon represents a possible target to develop preventive and therapeutic strategies against chronic and therapy-refractory infections.


Assuntos
Proteínas de Bactérias/metabolismo , Endotélio Vascular/microbiologia , Interações Hospedeiro-Patógeno , Neutrófilos/microbiologia , Osteoblastos/microbiologia , Fator sigma/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Adaptação Fisiológica , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Deleção de Genes , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Células Endoteliais da Veia Umbilical Humana/microbiologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Neutrófilos/citologia , Neutrófilos/imunologia , Neutrófilos/patologia , Osteoblastos/citologia , Osteoblastos/imunologia , Osteoblastos/patologia , Proteômica , Fator sigma/genética , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/patologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/metabolismo , Fatores de Tempo , Transativadores/genética , Transativadores/metabolismo
10.
Cell Microbiol ; 18(6): 807-19, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26595647

RESUMO

Mast cells (MCs) are important sentinels of the host defence against invading pathogens. We previously reported that Staphylococcus aureus evaded the extracellular antimicrobial activities of MCs by promoting its internalization within these cells via ß1 integrins. Here, we investigated the molecular mechanisms governing this process. We found that S. aureus responded to the antimicrobial mediators released by MCs by up-regulating the expression of α-hemolysin (Hla), fibronectin-binding protein A and several regulatory systems. We also found that S. aureus induced the up-regulation of ß1 integrin expression on MCs and that this effect was mediated by Hla-ADAM10 (a disintegrin and metalloproteinase 10) interaction. Thus, deletion of Hla or inhibition of Hla-ADAM10 interaction significantly impaired S. aureus internalization within MCs. Furthermore, purified Hla but not the inactive HlaH35L induced up-regulation of ß1 integrin expression in MCs in a dose-dependent manner. Our data support a model in which S. aureus counter-reacts the extracellular microbicidal mechanisms of MCs by increasing expression of fibronectin-binding proteins and by inducing Hla-ADAM10-mediated up-regulation of ß1 integrin in MCs. The up-regulation of bacterial fibronectin-binding proteins, concomitantly with the increased expression of its receptor ß1 integrin on the MCs, resulted in enhanced S. aureus internalization through the binding of fibronectin-binding proteins to integrin ß1 via fibronectin.


Assuntos
Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Integrina beta1/metabolismo , Mastócitos/microbiologia , Staphylococcus aureus/fisiologia , Staphylococcus aureus/patogenicidade , Proteína ADAM10/metabolismo , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Toxinas Bacterianas/genética , Células Cultivadas , Feminino , Regulação Bacteriana da Expressão Gênica , Proteínas Hemolisinas/genética , Interações Hospedeiro-Patógeno , Mastócitos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Infecções Cutâneas Estafilocócicas/metabolismo , Infecções Cutâneas Estafilocócicas/microbiologia , Regulação para Cima
11.
Curr Genet ; 62(1): 15-7, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26123224

RESUMO

Staphylococcus aureus is an important pathogen of severe invasive tissue infection, e.g. osteomyelitis that can develop to chronicity and become extremely difficult to treat. Recent research revealed that S. aureus can dynamically switch to small colony variants (SCVs) that are adapted bacterial phenotypes for long-term persistence. The underlying mechanisms of the bacterial switching and adaptation process are largely dependent on an intact Sigma B regulon. As SigB is known as a transcription factor that modulates the stress response of several Gram-positive bacteria, it is most likely required by the bacteria to cope with the intracellular stress conditions. Here, we demonstrate in a long-term infection model of human osteoblasts that S. aureus continuously upregulated the expression of SigB during intracellular persistence. The increased SigB expression was accompanied by upregulation of adhesins and downregulation of toxins, which are characteristics for SCV phenotypes. These data further stress the role of SigB during chronic infections that could be a novel target for preventive or therapeutic measures to avoid chronic infections.


Assuntos
Adaptação Biológica , Regulação Bacteriana da Expressão Gênica , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos , Fator sigma/genética , Fator sigma/metabolismo , Fatores de Virulência/genética
12.
Cell Microbiol ; 16(3): 364-77, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24118665

RESUMO

Binding to fibronectin (FN) is a crucial pathogenic factor of Staphylococcus aureus mediated by fibronectin-binding protein A (FnBP-A) and extracellular adherence protein (Eap). Recently, we have shown that binding of soluble CD163 (sCD163) to FN linked to these molecules exhibits anti-microbial effects by enhancing phagocytosis and killing activity of S. aureus-infected monocytes. However, it remained unclear whether sCD163 also influences the monocytic activation status. Using genetically modified staphylococcal strains we now identified FnBP-A, but not Eap, as activator of the inflammatory response of monocytes to infection. FnBP-A-mediated inflammatory activation was masked by sCD163 binding to S. aureus promoting efficient pathogen elimination. Thus, sCD163 protects monocytes from overwhelming activation upon staphylococcal infection by dampening the secretion of pro-inflammatory cytokines TNFα, IL-1ß, IL-6 and IL-8 and DAMP molecule MRP8/14. Moreover, sCD163 limited expression of pro-apoptotic transcription factor NR4A1 induced during S. aureus infection and inhibited induction of chemokine CXCL2promoting survival of staphylococci in vivo. sCD163-mediated effects were not due to general immunosuppression since MAP kinase activation and ROS production were unaltered during infection of monocytes with sCD163-bound bacteria. Thus, sCD163 promotes a specific defence of the immune system against FnBP-A-mediated inflammatory activation enabling successful pathogen elimination, tissue recovery and resolution of inflammation.


Assuntos
Adesinas Bacterianas/imunologia , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Monócitos/imunologia , Receptores de Superfície Celular/imunologia , Staphylococcus aureus/imunologia , Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Bactérias/imunologia , Calgranulina B/biossíntese , Células Cultivadas , Quimiocina CXCL2/biossíntese , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Interleucina-1beta/biossíntese , Interleucina-1beta/metabolismo , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Interleucina-8/biossíntese , Interleucina-8/metabolismo , Macrófagos/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/microbiologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/biossíntese , Fagocitose/imunologia , Proteínas de Ligação a RNA/imunologia , Espécies Reativas de Oxigênio/metabolismo , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/metabolismo
13.
Int J Med Microbiol ; 304(2): 170-6, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24365645

RESUMO

S. aureus is a frequent cause of chronic and therapy-refractory infections. The ability of S. aureus to invade different types of non-professional phagocytes, to escape from the host lysosomal degradation machinery and to persist within the intracellular location for long time periods are most likely essential steps in pathogenesis. During the course from acute to chronic infection the bacteria need to dynamically react to the environmental changes and to adapt to the intracellular environment. In this context the bacteria change to SCV-like phenotypes that exhibit some characteristics of stable SCV-mutants, like upregulation of adhesins and downregulation of toxins. The exact formation mechanism and further typical features of these dynamically forming SCVs are largely unknown. In this review, recent data on the essential steps to establish chronic infections will be summarized and the clinical consequences of the dynamic bacterial adaptation mechanisms will be discussed.


Assuntos
Fagócitos/imunologia , Fagócitos/microbiologia , Fagossomos/imunologia , Fagossomos/microbiologia , Staphylococcus aureus/fisiologia , Adaptação Fisiológica , Animais , Humanos , Evasão da Resposta Imune , Staphylococcus aureus/imunologia
14.
Int J Med Microbiol ; 304(8): 1038-49, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25129555

RESUMO

Osteomyelitis is a severe inflammatory disease of the bone that is mainly caused by Staphylococcus aureus. Particularly, bone infections are difficult to treat and can develop into a chronic course with a high relapsing rate despite of antimicrobial treatments. The complex interaction of staphylococci with osseous tissue and the bacterial ability to invade host cells are thought to determine the severity of infection. Yet, defined bacterial virulence factors responsible for the pathogenesis of osteomyelitis have not been clearly identified. The aim of this study was to detect S. aureus virulence factors that are associated with osteomyelitis and contribute to a chronic course of infection. To this purpose, we collected 41 S. aureus isolates, each 11 from acute osteomyelitis (infection period less than 2 months), 10 from chronic osteomyelitis (infection period more than 12 months), 10 from sepsis and 10 from nasal colonization. All isolates were analyzed for gene expression and in functional in-vitro systems. Adhesion assays to bone matrix revealed that all isolates equally bound to matrix structures, but invasion assays in human osteoblasts showed a high invasive capacity of chronic osteomyelitis isolates. The high invasion rate could not be explained by defined adhesins, as all infecting strains expressed a multitude of adhesins that act together and determine the level of adhesion. Following host cell invasion isolates from chronic osteomyelitis induced less cytotoxicity than all other isolates and a higher percentage of Small-colony-variant (SCV)-formation, which represents an adaptation mechanism during long-term persistence. Isolates from acute and chronic osteomyelitis strongly produced biofilm and highly expressed agr and sarA that regulate secreted virulence factors and induced an inflammatory response in osteoblasts. In conclusion, chronic osteomyelitis isolates were characterized by a high host cell invasion rate, low cytotoxicity and the ability to persist and adapt within osteoblasts. Furthermore, isolates from both acute and chronic osteomyelitis strongly produced biofilm and induced high levels of host cell inflammation, which may explain tissue destruction and bone deformation observed as typical complications of long-lasting bone infections.


Assuntos
Inflamação , Osteomielite/microbiologia , Osteomielite/patologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/fisiologia , Adaptação Fisiológica , Aderência Bacteriana , Doença Crônica , Endocitose , Interações Hospedeiro-Patógeno , Humanos , Staphylococcus aureus/isolamento & purificação , Virulência
15.
BMC Biol ; 11: 63, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23714179

RESUMO

BACKGROUND: Different non-invasive real-time imaging techniques have been developed over the last decades to study bacterial pathogenic mechanisms in mouse models by following infections over a time course. In vivo investigations of bacterial infections previously relied mostly on bioluminescence imaging (BLI), which is able to localize metabolically active bacteria, but provides no data on the status of the involved organs in the infected host organism. In this study we established an in vivo imaging platform by magnetic resonance imaging (MRI) for tracking bacteria in mouse models of infection to study infection biology of clinically relevant bacteria. RESULTS: We have developed a method to label Gram-positive and Gram-negative bacteria with iron oxide nano particles and detected and pursued these with MRI. The key step for successful labeling was to manipulate the bacterial surface charge by producing electro-competent cells enabling charge interactions between the iron particles and the cell wall. Different particle sizes and coatings were tested for their ability to attach to the cell wall and possible labeling mechanisms were elaborated by comparing Gram-positive and -negative bacterial characteristics. With 5-nm citrate-coated particles an iron load of 0.015 ± 0.002 pg Fe/bacterial cell was achieved for Staphylococcus aureus. In both a subcutaneous and a systemic infection model induced by iron-labeled S. aureus bacteria, high resolution MR images allowed for bacterial tracking and provided information on the morphology of organs and the inflammatory response. CONCLUSION: Labeled with iron oxide particles, in vivo detection of small S. aureus colonies in infection models is feasible by MRI and provides a versatile tool to follow bacterial infections in vivo. The established cell labeling strategy can easily be transferred to other bacterial species and thus provides a conceptual advance in the field of molecular MRI.


Assuntos
Escherichia coli/metabolismo , Imageamento por Ressonância Magnética , Staphylococcus aureus/metabolismo , Animais , Modelos Animais de Doenças , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Compostos Férricos/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/microbiologia , Humanos , Ferro/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Microscopia de Fluorescência , Nanopartículas/química , Fagocitose/efeitos dos fármacos , Reprodutibilidade dos Testes , Coloração e Rotulagem , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/ultraestrutura
16.
ISME Commun ; 4(1): ycae077, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38962494

RESUMO

Staphylococcus aureus (Sa) and Acinetobacter baumannii (Ab) are frequently co-isolated from polymicrobial infections that are severe and refractory to therapy. Here, we apply a combination of wet-lab experiments and in silico modeling to unveil the intricate nature of the Ab/Sa interaction using both, representative laboratory strains and strains co-isolated from clinical samples. This comprehensive methodology allowed uncovering Sa's capability to exert a partial interference on Ab by the expression of phenol-soluble modulins. In addition, we observed a cross-feeding mechanism by which Sa supports the growth of Ab by providing acetoin as an alternative carbon source. This study is the first to dissect the Ab/Sa interaction dynamics wherein competitive and cooperative strategies can intertwine. Through our findings, we illuminate the ecological mechanisms supporting their coexistence in the context of polymicrobial infections. Our research not only enriches our understanding but also opens doors to potential therapeutic avenues in managing these challenging infections.

17.
Am J Pathol ; 181(4): 1206-14, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22902429

RESUMO

Osteomyelitis is a serious bone infection typically caused by Staphylococcus aureus. The pathogenesis of osteomyelitis remains poorly understood, mainly for lack of experimental models that closely mimic human disease. We describe a novel murine model of metastatic chronic osteomyelitis initiated after intravenous inoculation of S. aureus microorganisms. The bacteria entered bones through the bloodstream and, after an acute phase with progressive growth (first 2 weeks after infection), they remained at constant numbers for up to 56 days (chronic phase). Clinical signs of illness and systemic inflammation were apparent only during the acute phase. Bone destruction and remodeling processes were readily detectable by magnetic resonance and X-ray imaging 3 weeks after infection, and high levels of bone deformation were observed during the chronic phase. Histological examination of infected bones demonstrated suppurative inflammation with foci of intense bacterial multiplication and necrosis during acute infection and osteoclastic resorption accompanied by new woven bone formation during chronic infection. Transmission electron microscopy revealed S. aureus microorganisms forming microcolonies within the nonmineralized collagen matrix or located intracellularly within neutrophils. In summary, our mouse model of staphylococcal hematogenous osteomyelitis precisely reproduces most features of the human disease. Although the extent of lesions in the chronic phase was subject to variation, this model is ideal for testing and monitoring novel treatment modalities via noninvasive imaging.


Assuntos
Osteomielite/microbiologia , Osteomielite/patologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/fisiologia , Animais , Fenômenos Biomecânicos , Doença Crônica , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Úmero/diagnóstico por imagem , Úmero/microbiologia , Úmero/patologia , Imageamento Tridimensional , Inflamação/patologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Osteomielite/fisiopatologia , Radiografia , Infecções Estafilocócicas/fisiopatologia , Tíbia/diagnóstico por imagem , Tíbia/microbiologia , Tíbia/patologia , Tíbia/ultraestrutura , Fatores de Tempo
18.
J Infect Dis ; 206(7): 1138-48, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22837490

RESUMO

Staphylococcus aureus necrotizing pneumonia is a life-threatening disease that is frequently preceded by influenza infection. The S. aureus toxin Panton-Valentine leukocidin (PVL) is most likely causative for necrotizing diseases, but the precise pathogenic mechanisms of PVL and a possible contribution of influenza virus remain to be elucidated. In this study, we present a model that explains how influenza virus and PVL act together to cause necrotizing pneumonia: an influenza infection activates the lung epithelium to produce chemoattractants for neutrophils. Upon superinfection with PVL-expressing S. aureus, the recruited neutrophils are rapidly killed by PVL, resulting in uncontrolled release of neutrophil proteases that damage the airway epithelium. The host counteracts this pathogen strategy by generating PVL-neutralizing antibodies and by neutralizing the released proteases via protease inhibitors present in the serum. These findings explain why necrotizing infections mainly develop in serum-free spaces (eg, pulmonary alveoli) and open options for new therapeutic approaches.


Assuntos
Toxinas Bacterianas/farmacologia , Epitélio/virologia , Exotoxinas/farmacologia , Vírus da Influenza A/fisiologia , Leucocidinas/farmacologia , Pulmão/patologia , Staphylococcus aureus/imunologia , Animais , Toxinas Bacterianas/metabolismo , Extratos Celulares/farmacologia , Linhagem Celular , Meios de Cultivo Condicionados , Epitélio/imunologia , Exotoxinas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/imunologia , Influenza Humana/complicações , Influenza Humana/imunologia , Influenza Humana/patologia , Leucocidinas/metabolismo , Pulmão/imunologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Ativação de Neutrófilo , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/virologia , Peptídeo Hidrolases/metabolismo , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/patologia , Inibidores de Proteases/farmacologia , Staphylococcus aureus/metabolismo , Carga Viral
19.
Microorganisms ; 11(4)2023 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-37110443

RESUMO

Staphylococcus aureus evades antibiotic therapy and antimicrobial defenses by entering human host cells. Bacterial transcriptomic analysis represents an invaluable tool to unravel the complex interplay between host and pathogen. Therefore, the extraction of high-quality RNA from intracellular S. aureus lays the foundation to acquire meaningful gene expression data. In this study, we present a novel and straightforward strategy to isolate RNA from internalized S. aureus after 90 min, 24 h, and 48 h postinfection. Real-time PCR data were obtained for the target genes agrA and fnba, which play major roles during infection. The commonly used reference genes gyrB, aroE, tmRNA, gmk, and hu were analyzed under different conditions: bacteria from culture (condition I), intracellular bacteria (condition II), and across both conditions I and II. The most stable reference genes were used for the normalization of agrA and fnbA. Delta Cq (quantification cycle) values had a relatively low variability and thus demonstrated the high quality of the extracted RNA from intracellular S. aureus during the early phase of infection. The established protocol allows the extraction and purification of intracellular staphylococcal RNA while minimizing the amount of host RNA in the sample. This approach can leverage reproducible gene expression data to study host-pathogen interactions.

20.
Biology (Basel) ; 11(4)2022 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-35453769

RESUMO

Polymicrobial infections are more challenging to treat and are recognized as responsible for significant morbidity and mortality. It has been demonstrated that multiple Gram-negative organisms take advantage of the effects of Staphylococcus aureus α-toxin on mucosal host defense, resulting in proliferation and dissemination of the co-infecting pathogens. Through phenotypic approaches, we observed a decrease in the motility of A. baumannii A118 after exposure to cell-free conditioned media (CFCM) of S. aureus strains, USA300 and LS1. However, the motility of A. baumannii A118 was increased after exposure to the CFCM of S. aureus strains USA300 Δhla and S. aureus LSI ΔagrA. Hemolytic activity was seen in A118, in the presence of CFCM of S. aureus LS1. Further, A. baumannii A118 showed an increase in biofilm formation and antibiotic resistance to tetracycline, in the presence of CFCM of S. aureus USA300. Transcriptomic analysis of A. baumannii A118, with the addition of CFCM from S. aureus USA300, was carried out to study A. baumannii response to S. aureus' released molecules. The RNA-seq data analysis showed a total of 463 differentially expressed genes, associated with a wide variety of functions, such as biofilm formation, virulence, and antibiotic susceptibility, among others. The present results showed that A. baumannii can sense and respond to molecules secreted by S. aureus. These findings demonstrate that A. baumannii may perceive and respond to changes in its environment; specifically, when in the presence of CFCM from S. aureus.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA