Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Gut Microbes ; 16(1): 2316932, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38356294

RESUMO

Mitochondrial dynamics are critical in cellular energy production, metabolism, apoptosis, and immune responses. Pathogenic bacteria have evolved sophisticated mechanisms to manipulate host cells' mitochondrial functions, facilitating their proliferation and dissemination. Salmonella enterica serovar Typhimurium (S. Tm), an intracellular foodborne pathogen, causes diarrhea and exploits host macrophages for survival and replication. However, S. Tm-associated mitochondrial dynamics during macrophage infection remain poorly understood. In this study, we showed that within macrophages, S. Tm remodeled mitochondrial fragmentation to facilitate intracellular proliferation mediated by Salmonella invasion protein A (SipA), a type III secretion system effector encoded by Salmonella pathogenicity island 1. SipA directly targeted mitochondria via its N-terminal mitochondrial targeting sequence, preventing excessive fragmentation and the associated increase in mitochondrial reactive oxygen species, loss of mitochondrial membrane potential, and release of mitochondrial DNA and cytochrome c into the cytosol. Macrophage replication assays and animal experiments showed that mitochondria and SipA interact to facilitate intracellular replication and pathogenicity of S. Tm. Furthermore, we showed that SipA delayed mitochondrial fragmentation by indirectly inhibiting the recruitment of cytosolic dynamin-related protein 1, which mediates mitochondrial fragmentation. This study revealed a novel mechanism through which S. Tm manipulates host mitochondrial dynamics, providing insights into the molecular interplay that facilitates S. Tm adaptation within host macrophages.


Assuntos
Microbioma Gastrointestinal , Salmonella typhimurium , Animais , Salmonella typhimurium/metabolismo , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/metabolismo , Sorogrupo , Dinâmica Mitocondrial , Proteínas de Bactérias/metabolismo , Macrófagos/metabolismo , Proliferação de Células
2.
Microbiol Spectr ; 10(6): e0347022, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36453941

RESUMO

Environmental DNA uptake by certain bacteria and its integration into their genome create genetic diversity and new phenotypes. DNA processing protein A (DprA) is part of a multiprotein complex and facilitates the natural transformation (NT) phenotype in most bacteria. Deinococcus radiodurans, an extremely radioresistant bacterium, is efficient in NT, and its genome encodes nearly all of the components of the natural competence complex. Here, we have characterized the DprA protein of this bacterium (DrDprA) for the known characteristics of DprA proteins of other bacteria and the mechanisms underlying the DNA-RecA interaction. DrDprA has three domains. In vitro studies showed that purified recombinant DrDprA binds to both single-strand DNA (ssDNA) and double-strand DNA (dsDNA) and is able to protect ssDNA from nucleolytic degradation. DrDprA showed a strong interaction with DrRecA and facilitated RecA-catalyzed functions in vivo. Mutational studies identified DrDprA amino acid residues crucial for oligomerization, the interaction with DrRecA, and DNA binding. Furthermore, we showed that both oligomerization and DNA binding properties of DrDprA are integral to its support of the DrRecA-catalyzed strand exchange reaction (SER) in vitro. Together, these data suggested that DrDprA is largely structurally conserved with other DprA homologs but shows some unique structure-function features like the existence of an additional C-terminal Drosophila melanogaster Miasto-like protein 1 (DML1) domain, equal affinities for ssDNA and dsDNA, and the collective roles of oligomerization and DNA binding properties in supporting DrRecA functions. IMPORTANCE Bacteria can take up extracellular DNA (eDNA) by natural transformation (NT). Many bacteria, including Deinococcus radiodurans, have constitutive competence systems and can take up eDNA throughout their growth phase. DprA (DNA processing protein A) is a transformation-specific recombination mediator protein (RMP) that plays a role in bacterial NT, and the absence of this gene significantly reduces the transformation efficiencies of both chromosomal and plasmid DNA. NT helps bacteria survive under adverse conditions and contributes to genetic diversity in bacteria. The present work describes the characterization of DprA from D. radiodurans and will add to the existing knowledge of DprA biology.


Assuntos
Deinococcus , Animais , Deinococcus/genética , Deinococcus/metabolismo , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/metabolismo , Proteínas de Bactérias/metabolismo , Drosophila melanogaster , DNA/metabolismo , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , DNA Bacteriano/genética , DNA Bacteriano/metabolismo
3.
BMC Microbiol ; 22(1): 259, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-36303110

RESUMO

BACKGROUND: Surface layer protein A (SlpA), the primary outermost structure of Clostridioides difficile, plays an essential role in C. difficile pathogenesis, although its interaction with host intestinal cells are yet to be understood. The aim of this study was to investigate the effects of SlpA extracted from C. difficile on tight junction (TJ) proteins expression and induction of pro-inflammatory cytokines in human colon carcinoma cell line HT-29. SlpA was extracted from three toxigenic C. difficile clinical strains including RT126, RT001, RT084 as well as C. difficile ATCC 700057 as non-toxigenic strain. Cell viability was performed by MTT assay, and the mRNA expression of TJ proteins and inflammation-associated genes was determined using quantitative RT-PCR. Additionally, the secretion of IL-8, IL-1ß and TNF-α cytokines was measured by ELISA. RESULTS: C. difficile SlpA from selected RTs variably downregulated the expression level of TJs-assassinated genes and increased the expression level of TLR-4 and pro-inflammatory cytokines in HT-29 treated cells. SlpA from RT126 significantly (padj<0.05) decreased the gene expression level of claudins family and JAM-A and increased the secretion of IL-8, TNF-α and IL1-ß as compared to untreated cells. Moreover, only SlpA from RT001 could significantly induce the expression of IL-6 (padj<0.05). CONCLUSION: The results of the present study highlighted the importance of SlpA in the pathogenesis of CDI and C. difficile-induced inflammatory response in the gut. Further studies are required to unravel the significance of the observed results in promoting the intestinal inflammation and immune response induced by C. difficile SlpA from different RTs.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Humanos , Ribotipagem , Clostridioides difficile/genética , Clostridioides , Proteína Estafilocócica A/genética , Junções Íntimas/metabolismo , Fator de Necrose Tumoral alfa/genética , Interleucina-8/genética , Proteínas de Bactérias/metabolismo , Células Epiteliais/metabolismo , Inflamação , Expressão Gênica
4.
Int Immunol ; 32(2): 89-104, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-31713625

RESUMO

Staphylococcus aureus is a main pathogen of osteomyelitis and protein A is a virulence factor with high affinity for IgG. In this study, we investigated whether S. aureus affects the differentiation and bone resorption of osteoclasts through the IgG-binding capacity of protein A. Staphylococcus aureus pre-treated with serum or IgG showed marked enhancement in osteoclastogenesis and bone resorption compared to non-treated S. aureus or a protein A-deficient mutant. Blocking of the Fc receptor and deletion of the Fcγ receptor gene in osteoclast precursor cells showed that enhanced osteoclastogenesis stimulated by S. aureus IgG immune complexes (ICs) was mediated by the Fc receptor on osteoclast precursor cells. In addition, osteoclastogenesis stimulated by S. aureus ICs but not the protein A-deficient mutant was markedly reduced in osteoclast precursor cells of Myd88-knockout mice. Moreover, NFATc1, Syk and NF-κB signals were necessary for osteoclastogenesis stimulated by S. aureus ICs. The results suggest the contribution of a of Toll-like receptor 2 (TLR2)-Myd88 signal to the activity of S. aureus ICs. We further examined the expression of pro-inflammatory cytokines that is known to be enhanced by FcγR-TLR cross-talk. Osteoclasts induced by S. aureus ICs showed higher expression of TNF-α and IL-1ß, and marked stimulation of proton secretion of osteoclasts activated by pro-inflammatory cytokines. Finally, injection of S. aureus, but not the protein A-deficient mutant, exacerbated bone loss in implantation and intra-peritoneal administration mouse models. Our results provide a novel mechanistic aspect of bone loss induced by S. aureus in which ICs and both Fc receptors and TLR pathways are involved.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Diferenciação Celular , Imunoglobulina G/imunologia , Receptores Fc/imunologia , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia , Receptor 2 Toll-Like/imunologia , Animais , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/imunologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoclastos/efeitos dos fármacos , Osteoclastos/imunologia , Osteogênese/efeitos dos fármacos , Ligante RANK/antagonistas & inibidores , Ligante RANK/farmacologia , Receptores Fc/deficiência , Receptores Fc/genética , Proteína Estafilocócica A/genética , Staphylococcus aureus/citologia , Ácidos Teicoicos/farmacologia
5.
Protein Expr Purif ; 166: 105520, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31644959

RESUMO

An affibody is a 58 amino acids peptide derived from the Z domain of staphylococcal protein A and generally applied in areas such as imaging diagnosis, clinical therapeutics and biotechnology research. To screen for an affibody targeting the immune checkpoint PD-L1, a combinatorial affibody library was generated in yeast using degenerate overlap PCR primers and In-fusion technology. Z-j1 and Z-j2 affibodies targeting the Ig-like V domain of PD-L1 were screened and identified from this combinatorial library using the yeast two hybrid system. The Z-j1 and Z-j2 recombinant affibody proteins were over produced in E.coli and purified. ELISA and GST pull-down assays showed that recombinant Z-j1 and Z-j2 affibody proteins bound with high affinity to PD-L1 and inhibited the interaction of PD-1/PD-L1. Thus, novel affibodies targeting the immune checkpoint PD-1/PD-L1 were identified and produced in this study and have the potential to be used in cancer immunotherapy.


Assuntos
Antineoplásicos/química , Antígeno B7-H1/imunologia , Peptídeos/genética , Receptor de Morte Celular Programada 1/imunologia , Proteínas Recombinantes de Fusão/genética , Sequência de Aminoácidos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Escherichia coli , Imunoterapia , Biblioteca de Peptídeos , Peptídeos/química , Domínios Proteicos , Proteínas Recombinantes de Fusão/química , Proteína Estafilocócica A/química , Proteína Estafilocócica A/genética
6.
Protein Expr Purif ; 152: 56-63, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30030046

RESUMO

Chaperone proteins are required to maintain the overall fold and function of proteins in the cell. As part of the Hsp70 family, Ssa1 acts to maintain cellular proteostasis through a variety of diverse pathways aimed to preserve the native conformation of target proteins, thereby preventing aggregation and future states of cellular toxicity. Studying the structural dynamics of Ssa1 in vitro is essential to determining their precise mechanisms and requires the development of purification methods that result in highly pure chaperones. Current methods of expressing and purifying Ssa1 utilize affinity tagged constructs expressed in Escherichia coli, however, expression in an exogenous source produces proteins that lack post-translational modifications leading to undesired structural and functional effects. Current protocols to purify Ssa1 from Saccharomyces cerevisiae require large amounts of starting material, multiple steps of chromatography, and result in low yield. Our objective was to establish a small-scale purification of Ssa1 expressed from its endogenous source, Saccharomyces cerevisiae, with significant yield and purity. We utilized a protein A affinity tag that was previously used to purify large protein complexes from yeast, combined with magnetic Dynabeads that are conjugated with rabbit immunoglobulin G (IgG). Our results show that we can produce native, highly pure, active Ssa1 via this one-step purification with minimal amounts of starting material, and this Ssa1-protein A fusion does not alter cellular phenotypes. This methodology is a significant improvement in Ssa1 purification and will facilitate future experiments that will elucidate the biochemical and biophysical properties of Hsp70 chaperones.


Assuntos
Adenosina Trifosfatases/isolamento & purificação , Biotecnologia/métodos , Proteínas de Choque Térmico HSP70/isolamento & purificação , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas de Saccharomyces cerevisiae/isolamento & purificação , Saccharomyces cerevisiae/genética , Proteína Estafilocócica A/isolamento & purificação , Adenosina Trifosfatases/biossíntese , Adenosina Trifosfatases/genética , Animais , Cromatografia de Afinidade/métodos , Clonagem Molecular , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP70/genética , Imunoglobulina G/química , Separação Imunomagnética/métodos , Coelhos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimologia , Proteínas de Saccharomyces cerevisiae/biossíntese , Proteínas de Saccharomyces cerevisiae/genética , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/metabolismo
8.
Virulence ; 9(1): 390-401, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29157101

RESUMO

Staphylococcus pseudintermedius is an opportunistic pathogen in dogs and the most frequent cause of canine pyoderma. Protein A, a potent virulence factor in S. aureus is encoded by the spa gene. S. pseudintermedius possesses genes seemingly analogous to spa, but the expression and the characteristics of their products have not been directly determined. The purpose of this study was to test isolates from major clonal groups for the presence of spa gene orthologs, quantitate their expression levels, and to characterize protein A in S. pseudintermedius. From the data, it was observed that S. pseudintermedius isolates express genes analogous to spa in S. aureus. Isolates representing major clonal populations in the United States and Europe, ST68 and ST71 respectively, bound significantly higher amounts of canine IgG than isolates with other genetic backgrounds, suggesting that these isolates have a higher density of protein A on their surface. Also, canine IgG bound to protein A on S. pseudintermedius via its Fc region similar to protein A from S. aureus. The mRNA profile differed based on the bacterial sequence types and correlated to the density of protein A on the bacterial surface. Protein A was also found to be secreted during the exponential growth phase. Phagocytosis experiments with S. pseudintermedius show that blocking of protein A enhanced phagocytosis in whole blood, neutrophils and in DH82 canine macrophage-like cell line. Taken together, the results demonstrate that S. pseudintermedius produces protein A that shares S. aureus protein A's ability to bind the Fc region of immunoglobulins and may serve as a potential virulence factor by evading the host immune system.


Assuntos
Perfilação da Expressão Gênica , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/metabolismo , Staphylococcus/genética , Staphylococcus/metabolismo , Animais , Células Cultivadas , Cães , Europa (Continente) , Técnicas de Silenciamento de Genes , Imunoglobulina G/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Fagocitose , Ligação Proteica , RNA Mensageiro/análise , RNA Mensageiro/genética , Staphylococcus/imunologia , Estados Unidos
9.
Plant Biotechnol J ; 16(2): 404-414, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28640955

RESUMO

Purification is a bottleneck and a major cost factor in the production of antibodies. We set out to engineer a bifunctional fusion protein from two building blocks, Protein A and a hydrophobin, aiming at low-cost and scalable antibody capturing in solutions. Immunoglobulin-binding Protein A is widely used in affinity-based purification. The hydrophobin fusion tag, on the other hand, has been shown to enable purification by two-phase separation. Protein A was fused to two different hydrophobin tags, HFBI or II, and expressed transiently in Nicotiana benthamiana. The hydrophobins enhanced accumulation up to 35-fold, yielding up to 25% of total soluble protein. Both fused and nonfused Protein A accumulated in protein bodies. Hence, the increased yield could not be attributed to HFB-induced protein body formation. We also demonstrated production of HFBI-Protein A fusion protein in tobacco BY-2 suspension cells in 30 l scale, with a yield of 35 mg/l. Efficient partitioning to the surfactant phase confirmed that the fusion proteins retained the amphipathic properties of the hydrophobin block. The reversible antibody-binding capacity of the Protein A block was similar to the nonfused Protein A. The best-performing fusion protein was tested in capturing antibodies from hybridoma culture supernatant with two-phase separation. The fusion protein was able to carry target antibodies to the surfactant phase and subsequently release them back to the aqueous phase after a change in pH. This report demonstrates the potential of hydrophobin fusion proteins for novel applications, such as harvesting antibodies in solutions.


Assuntos
Anticorpos/metabolismo , Nicotiana/metabolismo , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas/metabolismo , Proteína Estafilocócica A/metabolismo , Proteínas de Plantas/genética , Plantas Geneticamente Modificadas/genética , Proteína Estafilocócica A/genética , Nicotiana/genética
10.
PLoS One ; 12(8): e0183913, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28859130

RESUMO

Protein A in Staphylococcus aureus is encoded by the spa (staphylococcal protein A) gene and binds to immunoglobulin (Ig). The S. aureus strain Wood 46 has been variously reported as protein A-deficient and/or spa negative and used as a control in animal models of staphylococcal infections. The results of this study indicate that Wood 46 has normal spa expression but transcribes very low levels of the srtA gene which encodes the sortase A (SrtA) enzyme. This is consistent with unique mutations in the srtA promoter. In this study, a low level of sortase A explains deficient anchoring of proteins with an LPXTG motif, such as protein A, fibrinogen-binding protein and fibronectin-binding proteins A and B on to the peptidoglycan cell wall. The activity of secreted protein A is an important consideration for use of Wood 46 in functional experiments and animal models.


Assuntos
Adesinas Bacterianas/genética , Aminoaciltransferases/genética , Proteínas de Bactérias/genética , Proteínas de Transporte/genética , Cisteína Endopeptidases/genética , Regulação Bacteriana da Expressão Gênica , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética , Adesinas Bacterianas/metabolismo , Motivos de Aminoácidos , Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Proteínas de Transporte/metabolismo , Parede Celular/química , Parede Celular/metabolismo , Cisteína Endopeptidases/metabolismo , Mutação , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/metabolismo , Transcrição Gênica
11.
J Vet Med Sci ; 79(8): 1323-1326, 2017 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-28690290

RESUMO

Rhodococcus equi is a facultative intracellular bacterium that can escape from bactericidal mechanisms associated with phagocytosis. Virulence-associated protein A (VapA), encoded on a virulence-associated plasmid, is essential for intracellular survival in macrophages, but its function is not known. Here, we show that the extracellular addition of recombinant glutathione S-transferase (GST)-VapA fusion protein rescued the intracellular replication defect of a mutant lacking the vapA gene. Furthermore, the virulence-plasmid-cured strain could also multiply to nearly wild-type levels by the addition of GST-VapA. The present data suggest that VapA can alter the intraphagocytic environment, thereby affecting its suitability for the growth of R. equi.


Assuntos
Rhodococcus equi/genética , Rhodococcus equi/patogenicidade , Proteína Estafilocócica A/genética , Fatores de Virulência , Virulência/genética , Infecções por Actinomycetales/microbiologia , Animais , Linhagem Celular , Deleção de Genes , Genes Bacterianos/genética , Glutationa Transferase , Macrófagos/microbiologia , Camundongos , Plasmídeos , Proteínas Recombinantes , Rhodococcus equi/crescimento & desenvolvimento
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(5): 601-605, 2017 May.
Artigo em Chinês | MEDLINE | ID: mdl-28502296

RESUMO

Objective To investigate the effect of recombinant Legionella pneumophila flagella protein A (rflaA) on the secretion of interleukin-6 (IL-6) and interleukin-1ß (IL-1ß) by RAW264.7 macrophage and the possible mechanism. Methods RAW264.7 cells were treated with 0.000, 0.125, 0.250, 0.500, 1.000, 2.000, 4.000 and 8.000 µg/mL rflaA to determine the EC50 of rflaA using CCK-8 assay. Secretion of IL-6 and IL-1ß were measured by ELISA at 24, 36 and 48 hours after treatment of the cells with 0.04, 0.08 and 0.16 µg/mL rflaA. At 6, 12, 24, 36 and 48 hours after treatment of the cells with 0.04, 0.08 and 0.16 µg/mL rflaA, the expressions of IL-6, IL-1ß, NOD-like receptor protein 3 (NLRP3) and caspase-1 mRNAs were detected by quantitative real-time PCR, and the expressions of NLRP3 and caspase-1 proteins were tested by Western blotting. Results RflaA enhanced the expressions of IL-6 and IL-1ß, and the higher concentration of rflaA was more potential. The expressions of IL-6 and IL-1ß reached peak when the cells were treated with 0.16 µg/mL rflaA for 36 hours. Treatment of RAW264.7 cells with rflaA promoted the expressions of IL-6 and IL-1ß, NLRP3 and caspase-1 mRNA, and 0.16 µg/mL rflaA was the most potential at 12 hours after treatment. Expressions of NLRP3 and caspase-1 protein increased after treatment with rflaA, and 0.16 µg/mL rflaA induced the highest expression of both proteins at 24 hours after treatment. Conclusion RflaA could enhance the secretion of IL-6 and IL-1ß by promoting the expressions of NLRP3 and caspase-1 in RAW264.7 cells.


Assuntos
Flagelos/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Legionella pneumophila/metabolismo , Proteínas Recombinantes/farmacologia , Proteína Estafilocócica A/farmacologia , Animais , Linhagem Celular , Legionella pneumophila/genética , Camundongos , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteína Estafilocócica A/genética
13.
Virology ; 507: 151-160, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28437636

RESUMO

Flock House virus (FHV) RNA can be trans-encapsidated, entirely in planta, by tobacco mosaic virus coat protein to form virus-like particles (VLPs). Vaccination with these VLPs leads to strong antigen expression in mice and immune-activation. We hypothesize that creating an additional cellular site for replication and/or trans-encapsidation might significantly improve the final output of trans-encapsidated product. FHV protein A was engineered to target the endoplasmic reticulum (ER) via a heterologous tobacco etch virus ER-targeting domain, and was expressed in cis or in trans relative to the replicating FHV RNA1. A strong increase in marker gene expression in plants was noted when ER-targeted protein A was supplied in trans. RNA fluorescence in situ hybridization revealed RNA1 replication in both the mitochondria and ER, and total RNA1 accumulation was increased. In support of our hypothesis, VLP yield was increased significantly by the addition of this single genetic component to the inoculum.


Assuntos
Retículo Endoplasmático/virologia , Nicotiana/virologia , Nodaviridae/fisiologia , Proteína Estafilocócica A/química , Proteína Estafilocócica A/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Replicação Viral , Mitocôndrias/virologia , Nodaviridae/química , Nodaviridae/genética , Doenças das Plantas/virologia , Domínios Proteicos , Transporte Proteico , RNA Viral/genética , RNA Viral/metabolismo , Proteína Estafilocócica A/genética , Proteínas Virais/genética
14.
Artigo em Inglês | MEDLINE | ID: mdl-28409124

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA), is one of the most prevalent clinical pathogens isolated from hospital settings, and has increasingly identified in community settings. In China, the SCCmecIII-ST239 strains are disseminated in different geographic regions, accounting for >75% of all MRSA isolates in some national studies. Here we characterized 150 non-duplicate MRSA isolates collected from February 2012 to May 2013 in a tertiary hospital in Suzhou, Eastern China, to explore the molecular epidemiology. All isolates were characterized by spa typing, SCCmec typing, and detection of genes encoding Panton-Valentine leukocidin (PVL) and toxic shock syndrome toxin (TSST-1). Representative genotypes were also subjected to multilocus sequence typing (MLST). Antibiotic susceptibility testing was performed using BD Phoenix™ Automated Microbiology System. Molecular typing identified 11 clonal complex (CC) and 28 spa types, with the CC5-spa t002 (29.3%) and CC239-spa t037 (14.7%) being the most prevalent. SCCmec types II, III, IV, and V were identified in 33.3, 21.3, 23.3, and 21.3% of all isolates, respectively. PVL genes (lukF/S-PV) were detected in 11.3% of all isolates and from 6 CCs (5, 8, 59, 88, 239, and 398). The TSST-1 gene (tst) was detected in 18.0% of the all isolates, predominantly in CC5 (96.3%). All the tst-1-positve CC5 isolates were spa t002. Eighteen patients died within 30 days of hospitalization, and the in-hospital 30-day mortality was 12.0%. Multivariable analysis showed that 60 years old (odds ratio [OR] = 7.2, P = 0.026), cancer diagnosis (OR = 9.6, P = 0.022), and MRSA isolate carriage of tst-1 (OR = 62.5, P < 0.001) were independent factors associated with 30-day mortality. Our study revealed unique MRSA dissemination patterns in our hospital in comparison to those of other regions in China. The finding that tst-1-positive CC5 strains were associated with higher mortality highlights the need for strict infection control measures in order to prevent further spread of these strains in our hospital, as well as others.


Assuntos
Toxinas Bacterianas/genética , Infecção Hospitalar/epidemiologia , Infecção Hospitalar/microbiologia , Enterotoxinas/genética , Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Infecções Estafilocócicas/epidemiologia , Infecções Estafilocócicas/microbiologia , Superantígenos/genética , Adulto , Idoso , China/epidemiologia , Exotoxinas/genética , Feminino , Genótipo , Humanos , Leucocidinas/genética , Masculino , Staphylococcus aureus Resistente à Meticilina/classificação , Staphylococcus aureus Resistente à Meticilina/genética , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Epidemiologia Molecular , Tipagem Molecular , Fatores de Risco , Proteína Estafilocócica A/genética , Análise de Sobrevida , Centros de Atenção Terciária
15.
BMC Biotechnol ; 16: 22, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26911296

RESUMO

BACKGROUND: We previously developed in vitro immunization based on a fusion protein containing the transcriptional transactivator (Tat) of human immunodeficiency virus and a double domain, called ZZ, derived from protein A of Staphylococcus aureus. In this approach, naïve human peripheral blood mononuclear cells (PBMCs) trigger a specific IgM antibody (Ab) response in the presence of ZZTat. In the present study, we attempted to raise a specific IgG Ab response. RESULTS: We found that PBMCs incubated with ZZTat and a mixture containing anti-CD40, IL4 and IL21 secrete anti-Tat IgG Abs in their supernatants, indicating that the cytokine cocktail provides an isotypic switch. Then, we deciphered the Tat determinant involved in the phenomenon and found that it is located in the region 22-57 and that, within this region, the cysteine-rich domain and the basic residues play a crucial role. Finally, we prepared a fusion protein containing a fragment derived from the NY-ESO-1 cancer/testis antigen (Ag) and showed that PBMCs incubated with ZZfNY-ESO-1Tat trigger a specific anti-fNY-ESO-1 IgG Ab response, which demonstrates the possibility of transferring immunizing ability to an Ag unrelated to Tat. CONCLUSION: Our ZZTat-based in vitro immunization approach that offers the possibility to raise an IgG Ab response against NY-ESO-1 might represent a valuable first stage for the generation of fully human IgG specific Abs.


Assuntos
Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Leucócitos Mononucleares/imunologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Células Cultivadas , Humanos , Proteínas Recombinantes de Fusão/genética , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/imunologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
16.
Microbiol Immunol ; 60(3): 148-59, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26786482

RESUMO

A novel benzimidazole molecule that was identified in a small-molecule screen and is known as antibiofilm compound 1 (ABC-1) has been found to prevent bacterial biofilm formation by multiple bacterial pathogens, including Staphylococcus aureus, without affecting bacterial growth. Here, the biofilm inhibiting ability of 156 µM ABC-1 was tested in various biofilm-forming strains of S. aureus. It was demonstrated that ABC-1 inhibits biofilm formation by these strains at micromolar concentrations regardless of the strains' dependence on Polysaccharide Intercellular Adhesin (PIA), cell wall-associated protein dependent or cell wall- associated extracellular DNA (eDNA). Of note, ABC-1 treatment primarily inhibited Protein A (SpA) expression in all strains tested. spa gene disruption showed decreased biofilm formation; however, the mutants still produced more biofilm than ABC-1 treated strains, implying that ABC-1 affects not only SpA but also other factors. Indeed, ABC-1 also attenuated the accumulation of PIA and eDNA on cell surface. Our results suggest that ABC-1 has pleotropic effects on several biofilm components and thus inhibits biofilm formation by S. aureus.


Assuntos
Antibacterianos/farmacologia , Benzimidazóis/farmacologia , Biofilmes/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , Aminoaciltransferases/genética , Antibacterianos/síntese química , Antibacterianos/química , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Benzimidazóis/química , Biofilmes/crescimento & desenvolvimento , Parede Celular/metabolismo , Cisteína Endopeptidases/genética , Regulação para Baixo , Polissacarídeos Bacterianos/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteína Estafilocócica A/biossíntese , Proteína Estafilocócica A/efeitos dos fármacos , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
17.
Klin Lab Diagn ; 61(8): 498-501, 2016 Aug.
Artigo em Russo | MEDLINE | ID: mdl-30601643

RESUMO

Staphylococcus aureus and Pseudomonas aeruginosa are foregroundpathogens of bacteriemia and sepsis. They produce large spectrum of such factors of pathogenicity permitting them to proliferate and survive in bloodstream as hydrolytic enzymes, adenosine diphosphate-ribosylarginine toxins, plasmocoagulase, etc. The occurrence of alteration of growth and expression of virulence factors of S. aureus and P. aeruginosa at fermentation in LB-broth depending on iron concentration was demonstrated previously. The conditions in vivo significantly differ the laboratory conditions. The activity of growth and expression of pathogenicity factors of S. aureus and P. aeruginosa at fermentation in blood serum of donors with different alternative of iron homeostasis was analyzed. The study established expression of genes of hemolytic phospholipase C (plcH), alginate (algD), exotoxin A (exoA) for P. aeruginosa and genes of protein A (spA), virulence global regulator (RNAIII) for S. aureus. The iron-deficient serum and serum with normal iron homeostasis decreased activity of growth of S. aureus and P. aeruginosa. The growth rate and expression level of all analyzed genes turned out higher at fermentation of S. aureus and P. aeruginosa in serums containing excess of iron (more than 30 mkM). The fermentation of P. aeruginosa in iron-deficient serum resulted in decreasing of expression level of genes plcH, algD, exoA. The expression of RNA III and spaA S.aureus in iron-deficient serum increased towards normal content of iron in blood. The example of S. aureus and P. aeruginosa demonstrated that expression of many virulence factors of opportunistic microorganisms depends on iron homeostasis of host organism. The survival and proliferation of microorganisms in blood depend on both immune status of host organism and biological characteristics of pathogen.


Assuntos
Anemia Ferropriva/sangue , Ferro/sangue , Infecções Oportunistas/microbiologia , Pseudomonas aeruginosa/genética , Staphylococcus aureus/genética , ADP Ribose Transferases/genética , Adulto , Anemia Ferropriva/microbiologia , Toxinas Bacterianas/genética , Proliferação de Células/efeitos dos fármacos , Exotoxinas/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Masculino , Infecções Oportunistas/genética , Infecções Oportunistas/patologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/patogenicidade , RNA Bacteriano/genética , Soro/química , Soro/microbiologia , Proteína Estafilocócica A/genética , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/patogenicidade , Transferases (Outros Grupos de Fosfato Substituídos)/genética , Fatores de Virulência/genética , Exotoxina A de Pseudomonas aeruginosa
18.
Eur J Clin Microbiol Infect Dis ; 34(9): 1833-7, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26076750

RESUMO

Staphylococcus aureus (S. aureus) is the most common cause of bone and joint infections. However, limited information is available on the distribution of S. aureus geno- and phenotypes causing orthopaedic infections. The aim of this study was to identify the dominating types causing infections in orthopaedic patients, investigate if the characteristics of these types changed over time and examine if different types were more often associated with surgical site infection (SSI) than primary infection (non-SSI). All clinical S. aureus isolates collected from orthopaedic patients from 2000 through 2011 at Akershus University Hospital, Norway, were characterised by S. aureus protein A (spa) typing and tested for antibiotic resistance. A total of 548 patients with orthopaedic S. aureus infections were included, of which 326 (59 %) had SSI and 222 (41 %) had non-SSI. The median age was 62 years [range 2-97 years] and 54 % were male. Among the 242 unique spa types, t084 was the most common (7 %). Penicillin resistance was identified in 75 % of the isolates, whereas the resistances to the other antibiotics tested were <5 %. Three isolates (0.5 %) were resistant to methicillin. There was no significant difference in the distribution of geno- and phenotypes over time and there was no difference in types between SSI and non-SSI. In this large collection of S. aureus from orthopaedic patients, the S. aureus infections, regardless of origin, were heterogeneous, mainly resistant to penicillin, stable over time and consisted of similar types as previously found in both carrier and other patient populations.


Assuntos
Doenças Ósseas Infecciosas/microbiologia , Artropatias/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/isolamento & purificação , Infecção da Ferida Cirúrgica/microbiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibacterianos/uso terapêutico , Técnicas de Tipagem Bacteriana , Doenças Ósseas Infecciosas/tratamento farmacológico , Criança , Pré-Escolar , Estudos de Coortes , Infecção Hospitalar/microbiologia , Farmacorresistência Bacteriana , Feminino , Humanos , Artropatias/tratamento farmacológico , Masculino , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Procedimentos Ortopédicos/efeitos adversos , Estudos Prospectivos , Infecções Estafilocócicas/microbiologia , Proteína Estafilocócica A/genética , Staphylococcus aureus/classificação , Staphylococcus aureus/efeitos dos fármacos , Infecção da Ferida Cirúrgica/tratamento farmacológico , Adulto Jovem
19.
Infect Immun ; 83(4): 1598-609, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25644005

RESUMO

The immunoglobulin binding protein A (SpA) of Staphylococcus aureus is synthesized as a precursor with a C-terminal sorting signal. The sortase A enzyme mediates covalent attachment to peptidoglycan so that SpA is displayed on the surface of the bacterium. Protein A is also found in the extracellular medium, but the processes involved in its release are not fully understood. Here, we show that a portion of SpA is released into the supernatant with an intact sorting signal, indicating that it has not been processed by sortase A. Release of SpA was reduced when the native sorting signal of SpA was replaced with the corresponding region of another sortase-anchored protein (SdrE). Similarly, a reporter protein fused to the sorting signal of SpA was released to a greater extent than the same polypeptide fused to the SdrE sorting signal. Released SpA protected bacteria from killing in human blood, indicating that it contributes to immune evasion.


Assuntos
Aminoaciltransferases/imunologia , Proteínas de Bactérias/imunologia , Parede Celular/imunologia , Cisteína Endopeptidases/imunologia , Evasão da Resposta Imune/imunologia , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia , Aminoaciltransferases/biossíntese , Aminoaciltransferases/genética , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/biossíntese , Cisteína Endopeptidases/genética , Endopeptidases/metabolismo , Humanos , Peptidoglicano/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Proteína Estafilocócica A/biossíntese , Proteína Estafilocócica A/genética , Staphylococcus aureus/metabolismo
20.
Intervirology ; 58(6): 363-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26909539

RESUMO

Virus-like particle (VLP) of JC polyomavirus (JCPyV) is capable of packaging and delivering exogenous DNA into human cells and can be used for mediating therapeutic gene expression. However, many human cells express the JCPyV receptor. Thus, wild-type VLP can transduce a wide range of human cells nonspecifically. This study tested the feasibility of using a modified VLP with a IgG binding domain (Z domain) of protein A in its capsid for targeted gene delivery. The Z domain of protein A isolated from the membrane of Staphylococcus aureus was inserted into the NH3-terminus of VP1, the major JCPyV capsular protein. The recombinant VLP-Z was produced using Escherichia coli. Electron-microscopic analysis showed that VLP-Z has a viral-like structure. A hemagglutination test showed that VLP-Z has hemagglutination activity. VP(1) was detected in the nuclei of HeLa cells by immunostaining after VLP-Z inoculation, suggesting that VLP-Z is viable and can enter the cell nucleus. Inoculating HeLa cells with pEGFP-N(1) plasmid packaged in VLP-Z has resulted in enhanced green fluorescent protein expression in the cells. In addition, a binding assay showed that VLP-Z was able to bind IgG through the interaction of the Z domain in VLP-Z and IgG. These data suggest that VLP-Z could be armed with cell-specific antibody and be used to deliver therapeutic genes to target cells.


Assuntos
Imunoglobulina G/metabolismo , Vírus JC/genética , Staphylococcus aureus/genética , Proteínas do Capsídeo/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Marcação de Genes , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Vírus JC/ultraestrutura , Plasmídeos/genética , Proteína Estafilocócica A/genética , Staphylococcus aureus/metabolismo , Vírion/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA