Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
País como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Med Microbiol ; 314: 151610, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38310676

RESUMO

Shiga toxin-producing E. coli (STEC), including the subgroup of enterohemorrhagic E. coli (EHEC), are important bacterial pathogens which cause diarrhea and the severe clinical manifestation hemolytic uremic syndrome (HUS). Genomic surveillance of STEC/EHEC is a state-of-the-art tool to identify infection clusters and to extract markers of circulating clinical strains, such as their virulence and resistance profile for risk assessment and implementation of infection prevention measures. The aim of the study was characterization of the clinical STEC population in Germany for establishment of a reference data set. To that end, from 2020 to 2022 1257 STEC isolates, including 39 of known HUS association, were analyzed and lead to a classification of 30.4 % into 129 infection clusters. Major serogroups in all clinical STEC analyzed were O26, O146, O91, O157, O103, and O145; and in HUS-associated strains were O26, O145, O157, O111, and O80. stx1 was less frequently and stx2 or a combination of stx, eaeA and ehxA were more frequently found in HUS-associated strains. Predominant stx gene subtypes in all STEC strains were stx1a (24 %) and stx2a (21 %) and in HUS-associated strains were mainly stx2a (69 %) and the combination of stx1a and stx2a (12.8 %). Furthermore, two novel O-antigen gene clusters (RKI6 and RKI7) and strains of serovars O45:H2 and O80:H2 showing multidrug resistance were detected. In conclusion, the implemented surveillance tools now allow to comprehensively define the population of clinical STEC strains including those associated with the severe disease manifestation HUS reaching a new surveillance level in Germany.


Assuntos
Escherichia coli Êntero-Hemorrágica , Infecções por Escherichia coli , Proteínas de Escherichia coli , Síndrome Hemolítico-Urêmica , Escherichia coli Shiga Toxigênica , Humanos , Virulência/genética , Antígenos O/genética , Proteínas de Escherichia coli/genética , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/microbiologia , Genômica , Alemanha/epidemiologia , Síndrome Hemolítico-Urêmica/epidemiologia , Síndrome Hemolítico-Urêmica/microbiologia , Família Multigênica
2.
J Virol ; 88(14): 8102-15, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24807719

RESUMO

Despite its strong host tropism for erythroid progenitor cells, human parvovirus B19 (B19V) can also infect a variety of additional cell types. Acute and chronic inflammatory cardiomyopathies have been associated with a high prevalence of B19V DNA in endothelial cells of the myocardium. To elucidate the mechanisms of B19V uptake into endothelium, we first analyzed the surface expression of the well-characterized primary B19V receptor P antigen and the putative coreceptors α5ß1 integrins and Ku80 antigen on primary and permanent endothelial cells. The receptor expression pattern and also the primary attachment levels were similar to those in the UT7/Epo-S1 cell line regarded as functional for B19V entry, but internalization of the virus was strongly reduced. As an alternative B19V uptake mechanism in endothelial cells, we demonstrated antibody-dependent enhancement (ADE), with up to a 4,000-fold increase in B19V uptake in the presence of B19V-specific human antibodies. ADE was mediated almost exclusively at the level of virus internalization, with efficient B19V translocation to the nucleus. In contrast to monocytes, where ADE of B19V has been described previously, enhancement does not rely on interaction of the virus-antibody complexes with Fc receptors (FcRs), but rather, involves an alternative mechanism mediated by the heat-sensitive complement factor C1q and its receptor, CD93. Our results suggest that ADE represents the predominant mechanism of endothelial B19V infection, and it is tempting to speculate that it may play a role in the pathogenicity of cardiac B19V infection. Importance: Both efficient entry and productive infection of human parvovirus B19 (B19V) seem to be limited to erythroid progenitor cells. However, in vivo, the viral DNA can also be detected in additional cell types, such as endothelial cells of the myocardium, where its presence has been associated with acute and chronic inflammatory cardiomyopathies. In this study, we demonstrated that uptake of B19V into endothelial cells most probably does not rely on the classical receptor-mediated route via the primary B19V receptor P antigen and coreceptors, such as α5ß1 integrins, but rather on antibody-dependent mechanisms. Since the strong antibody-dependent enhancement (ADE) of B19V entry requires the CD93 surface protein, it very likely involves bridging of the B19V-antibody complexes to this receptor by the complement factor C1q, leading to enhanced endocytosis of the virus.


Assuntos
Anticorpos Antivirais/metabolismo , Anticorpos Facilitadores , Células Endoteliais/virologia , Glicoproteínas de Membrana/metabolismo , Parvovirus B19 Humano/fisiologia , Receptores de Complemento/metabolismo , Receptores Virais/metabolismo , Internalização do Vírus , Linhagem Celular , Voluntários Saudáveis , Humanos , Parvovirus B19 Humano/imunologia
3.
Mol Ther ; 21(5): 924-33, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23439498

RESUMO

Insertion of completely complementary microRNA (miR) target sites (miRTS) into a transgene has been shown to be a valuable approach to specifically repress transgene expression in non-targeted tissues. miR-122TS have been successfully used to silence transgene expression in the liver following systemic application of cardiotropic adeno-associated virus (AAV) 9 vectors. For miR-206-mediated skeletal muscle-specific silencing of miR-206TS-bearing AAV9 vectors, however, we found this approach failed due to the expression of another member (miR-1) of the same miR family in heart tissue, the intended target. We introduced single-nucleotide substitutions into the miR-206TS and searched for those which prevented miR-1-mediated cardiac repression. Several mutated miR-206TS (m206TS), in particular m206TS-3G, were resistant to miR-1, but remained fully sensitive to miR-206. All these variants had mismatches in the seed region of the miR/m206TS duplex in common. Furthermore, we found that some m206TS, containing mismatches within the seed region or within the 3' portion of the miR-206, even enhanced the miR-206- mediated transgene repression. In vivo expression of m206TS-3G- and miR-122TS-containing transgene of systemically applied AAV9 vectors was strongly repressed in both skeletal muscle and the liver but remained high in the heart. Thus, site-directed mutagenesis of miRTS provides a new strategy to differentiate transgene de-targeting of related miRs.


Assuntos
Dependovirus/fisiologia , Regulação da Expressão Gênica , Inativação Gênica , Vetores Genéticos/genética , MicroRNAs/genética , Músculo Esquelético/metabolismo , Transgenes , Pareamento de Bases , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Ordem dos Genes , Técnicas de Transferência de Genes , Genes Reporter , Vetores Genéticos/metabolismo , Humanos , MicroRNAs/metabolismo , Mutagênese Sítio-Dirigida , Miocárdio/metabolismo , Especificidade de Órgãos/genética , Tropismo Viral
4.
Circulation ; 120(23): 2358-66, 2009 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-19933937

RESUMO

BACKGROUND: Group B coxsackieviruses (CVBs) are the prototypical agents of acute myocarditis and chronic dilated cardiomyopathy, but an effective targeted therapy is still not available. Here, we analyze the therapeutic potential of a soluble (s) virus receptor molecule against CVB3 myocarditis using a gene therapy approach. METHODS AND RESULTS: We generated an inducible adenoviral vector (AdG12) for strict drug-dependent delivery of sCAR-Fc, a fusion protein composed of the coxsackievirus-adenovirus receptor (CAR) extracellular domains and the carboxyl terminus of human IgG1-Fc. Decoy receptor expression was strictly doxycycline dependent, with no expression in the absence of an inducer. CVB3 infection of HeLa cells was efficiently blocked by supernatant from AdG12-transduced cells, but only in the presence of doxycycline. After liver-specific transfer, AdG12 (plus doxycycline) significantly improved cardiac contractility and diastolic relaxation compared with a control vector in CVB3-infected mice if sCAR-Fc was induced before infection (left ventricular pressure 59+/-3.8 versus 45.4+/-2.7 mm Hg, median 59 versus 45.8 mm Hg, P<0.01; dP/dt(max) 3645.1+/-443.6 versus 2057.9+/-490.2 mm Hg/s, median 3526.6 versus 2072 mm Hg/s, P<0.01; and dP/dt(min) -2125.5+/-330.5 versus -1310.2+/-330.3 mm Hg/s, median -2083.7 versus -1295.9 mm Hg/s, P<0.01) and improved contractility if induced concomitantly with infection (left ventricular pressure 76.4+/-19.2 versus 56.8+/-10.3 mm Hg, median 74.8 versus 54.4 mm Hg, P<0.05; dP/dt(max) 5214.2+/-1786.2 versus 3011.6+/-918.3 mm Hg/s, median 5182.1 versus 3106.6 mm Hg/s, P<0.05), respectively. Importantly, hemodynamics of animals treated with AdG12 (plus doxycycline) were similar to uninfected controls. Preinfection induction of sCAR-Fc completely blocked and concomitant induction strongly reduced cardiac CVB3 infection, myocardial injury, and inflammation. CONCLUSIONS: AdG12-mediated sCAR-Fc delivery prevents cardiac dysfunction in CVB3 myocarditis under prophylactic and therapeutic conditions.


Assuntos
Infecções por Coxsackievirus/prevenção & controle , Regulação Viral da Expressão Gênica , Miocardite/prevenção & controle , Receptores Virais/biossíntese , Receptores Virais/genética , Doença Aguda , Animais , Cardiomiopatias/genética , Cardiomiopatias/prevenção & controle , Cardiomiopatias/virologia , Infecções por Coxsackievirus/genética , Terapia Genética/métodos , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/genética , Miocardite/virologia , Receptores Virais/administração & dosagem
5.
Vet Microbiol ; 200: 130-137, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26654217

RESUMO

The increase of Escherichia coli producing extended-spectrum ß-lactamases (ESBL) in hospitals and their emergence as intestinal colonisers of healthy humans is of concern. Transmission ways and the extent of spread of distinct E. coli clones or ESBL genes among humans and animals via the food chain or the environment is a matter of debate. In this study we determined ESBL genotypes in E. coli isolates (n=233) resistant to 3rd generation cephalosporins from hospitals and medical practices using PCR and sequencing. Bacterial strain typing was performed by PCR-based phylogrouping, multilocus sequence typing (MLST) and a ST131-specific PCR. Results showed that CTX-M-15 (50.4%), CTX-M-1 (28.4%) and CTX-M-14 (5.6%) were the most common ESBL types. Especially, CTX-M-15 was associated with E. coli ST131 of phylogenetic group B2, which was the dominant sequence type among our isolates (35.8%). MLST typing revealed 40 different sequence types (STs), with ST131, ST410, ST10 and ST38 as the most prevalent ones. Our findings give an overview of the current distribution of ESBL-producing E. coli isolates from humans in Germany. E. coli O25b:H4-ST131 was confirmed to be the most common clone, which is known for its successful dissemination worldwide. Although heterogeneity among the isolates was found, several successful clones previously described in animals (ST410, ST10) also occurred in our isolate collection. Further detailed investigations of ESBL-producing isolates from different habitats are needed to evaluate possible transfer ways.


Assuntos
Resistência a Múltiplos Medicamentos/genética , Infecções por Escherichia coli/microbiologia , Escherichia coli/genética , beta-Lactamases/genética , Instituições de Assistência Ambulatorial , Animais , Técnicas de Tipagem Bacteriana , Escherichia coli/enzimologia , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/epidemiologia , Genótipo , Alemanha/epidemiologia , Hospitais , Humanos , Epidemiologia Molecular , Tipagem de Sequências Multilocus , Filogenia , Prevalência
6.
PLoS One ; 9(3): e92188, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24670775

RESUMO

In failing rat hearts, post-transcriptonal inhibition of phospholamban (PLB) expression by AAV9 vector-mediated cardiac delivery of short hairpin RNAs directed against PLB (shPLBr) improves both impaired SERCA2a controlled Ca2+ cycling and contractile dysfunction. Cardiac delivery of shPLB, however, was reported to cause cardiac toxicity in canines. Thus we developed a new AAV vector, scAAV6-amiR155-PLBr, expressing a novel engineered artificial microRNA (amiR155-PLBr) directed against PLB under control of a heart-specific hybrid promoter. Its PLB silencing efficiency and safety were compared with those of an AAV vector expressing shPLBr (scAAV6-shPLBr) from an ubiquitously active U6 promoter. Investigations were carried out in cultured neonatal rat cardiomyocytes (CM) over a period of 14 days. Compared to shPLBr, amiR155-PLBr was expressed at a significantly lower level, resulting in delayed and less pronounced PLB silencing. Despite decreased knockdown efficiency of scAAV6-amiR155-PLBr, a similar increase of the SERCA2a-catalyzed Ca2+ uptake into sarcoplasmic reticulum (SR) vesicles was observed for both the shPLBr and amiR155-PLBr vectors. Proteomic analysis confirmed PLB silencing of both therapeutic vectors and revealed that shPLBr, but not the amiR155-PLBr vector, increased the proinflammatory proteins STAT3, STAT1 and activated STAT1 phosphorylation at the key amino acid residue Tyr701. Quantitative RT-PCR analysis detected alterations in the expression of several cardiac microRNAs after treatment of CM with scAAV6-shPLBr and scAAV6-amiR155-PLBr, as well as after treatment with its related amiR155- and shRNAs-expressing control AAV vectors. The results demonstrate that scAAV6-amiR155-PLBr is capable of enhancing the Ca2+ transport function of the cardiac SR PLB/SERCA2a system as efficiently as scAAV6-shPLBr while offering a superior safety profile.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Dependovirus/metabolismo , Inativação Gênica , Vetores Genéticos/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Western Blotting , DNA Complementar/genética , Genes Reporter , Células HEK293 , Humanos , Inflamação/patologia , Miocárdio/metabolismo , Proteômica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Reprodutibilidade dos Testes , Retículo Sarcoplasmático , Transdução Genética
7.
Antiviral Res ; 88(1): 86-94, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20708037

RESUMO

Adenoviruses are pathological agents inducing mild respiratory and gastrointestinal infections. Under certain circumstances, for example in immunosuppressed patients, they induce severe infections of the liver, heart and lung, sometimes leading to death. Currently, adenoviral infections are treated by palliative care with no curative antiviral therapy yet available. Gene silencing by RNA interference (RNAi) has been shown to be a potent new therapeutic option for antiviral therapy. In the present study, we examined the potential of RNAi-mediated inhibition of adenovirus 5 infection by the use of small interfering (si)RNAs targeting both early (E1A) and late (hexon, IVa2) adenoviral genes. Several of the initially analyzed siRNAs directed against E1A, hexon and IVa2 showed a distinct antiviral activity. Among them, one siRNA for each gene was selected and used for the further comparative investigations of their efficiency to silence adenoviruses. Silencing of the late genes was more efficient in inhibiting adenoviral replication than comparable silencing of the E1A early gene. A combination strategy involving down-regulation of any two or all three of the targeted genes did not result in an enhanced inhibition of viral replication as compared to the single siRNA approaches targeting the late genes. However, protection against adenovirus-mediated cytotoxicity was substantially improved by combining siRNAs against either of the two late genes with the siRNA against the E1A early gene. Thus, an enhanced anti-adenoviral efficiency of RNAi-based inhibition strategies can be achieved by co-silencing of early and late adenoviral genes, with down regulation of the E1A as a crucial factor.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Virais/genética , Adenoviridae/fisiologia , Infecções por Adenovirus Humanos/terapia , Infecções por Adenovirus Humanos/virologia , Adenovírus Humanos/genética , Adenovírus Humanos/fisiologia , Antivirais , Células Cultivadas , Regulação para Baixo , Técnica Indireta de Fluorescência para Anticorpo , Genes Virais , Células HeLa , Humanos , Luciferases , Reação em Cadeia da Polimerase , RNA de Cadeia Dupla/efeitos dos fármacos , RNA Viral/genética , Transcrição Gênica , Ensaio de Placa Viral , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa