Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Microbiol Spectr ; 12(1): e0291223, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38084982

RESUMO

IMPORTANCE: Streptococcus pneumoniae (Spn) colonizes the lungs, killing millions every year. During its metabolism, Spn produces abundant amounts of hydrogen peroxide. When produced in the lung parenchyma, Spn-hydrogen peroxide (H2O2) causes the death of lung cells, and details of the mechanism are studied here. We found that Spn-H2O2 targets intracellular proteins, resulting in the contraction of the cell cytoskeleton and disruption of mitochondrial function, ultimately contributing to cell death. Intracellular proteins targeted by Spn-H2O2 included cytochrome c and, surprisingly, a protein of the cell cytoskeleton, beta-tubulin. To study the details of oxidative reactions, we used, as a surrogate model, the oxidation of another hemoprotein, hemoglobin. Using the surrogate model, we specifically identified a highly reactive radical whose creation was catalyzed by Spn-H2O2. In sum, we demonstrated that the oxidation of intracellular targets by Spn-H2O2 plays an important role in the cytotoxicity caused by Spn, thus providing new targets for interventions.


Assuntos
Peróxido de Hidrogênio , Streptococcus pneumoniae , Humanos , Streptococcus pneumoniae/metabolismo , Peróxido de Hidrogênio/toxicidade , Peróxido de Hidrogênio/metabolismo , Pulmão/metabolismo , Mitocôndrias/metabolismo , Respiração , Citoesqueleto/metabolismo
2.
Microbiol Spectr ; 12(1): e0329723, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38038454

RESUMO

IMPORTANCE: Heme degradation provides pathogens with growth essential iron, leveraging on the host heme reservoir. Bacteria typically import and degrade heme enzymatically, and here, we demonstrated a significant deviation from this dogma. We found that Streptococcus pneumoniae liberates iron from met-hemoglobin extracellularly, in a hydrogen peroxide (H2O2)- and cell-dependent manner; this activity serves as a major iron acquisition mechanism for S. pneumoniae. Inhabiting oxygen-rich environments is a major part of pneumococcal biology, and hence, H2O2-mediated heme degradation likely supplies iron during infection. Moreover, H2O2 reaction with ferrous hemoglobin but not with met-hemoglobin is known to result in heme breakdown. Therefore, the ability of pneumococci to degrade heme from met-hemoglobin is a new paradigm. Lastly, this study will inform other research as it demonstrates that extracellular degradation must be considered in the interpretations of experiments in which H2O2-producing bacteria are given heme or hemoproteins as an iron source.


Assuntos
Peróxido de Hidrogênio , Streptococcus pneumoniae , Peróxido de Hidrogênio/metabolismo , Streptococcus pneumoniae/metabolismo , Hemoglobinas/metabolismo , Heme/metabolismo , Ferro/metabolismo
3.
bioRxiv ; 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38014009

RESUMO

Pneumococcal pneumonia causes cytotoxicity in the lung parenchyma but the underlying mechanism involves multiple factors contributing to cell death. Here, we discovered that hydrogen peroxide produced by Streptococcus pneumoniae (Spn-H 2 O 2 ) plays a pivotal role by oxidizing hemoglobin, leading to its polymerization and subsequent release of labile heme. At physiologically relevant levels, heme selected a population of encapsulated pneumococci. In the absence of capsule and Spn-H 2 O 2 , host intracellular heme exhibited toxicity towards pneumococci, thus acting as an antibacterial mechanism. Further investigation revealed that heme-mediated toxicity required the ABC transporter GlnPQ. In vivo experiments demonstrated that pneumococci release H 2 O 2 to cause cytotoxicity in bronchi and alveoli through the non-proteolytic degradation of intracellular proteins such as actin, tubulin and GAPDH. Overall, our findings uncover a mechanism of lung toxicity mediated by oxidative stress that favor the growth of encapsulated pneumococci suggesting a therapeutic potential by targeting oxidative reactions. Highlights: Oxidation of hemoglobin by Streptococcus pneumoniae facilitates differentiation to encapsulated pneumococci in vivo Differentiated S. pneumoniae produces capsule and hydrogen peroxide (Spn-H 2 O 2 ) as defense mechanism against host heme-mediated toxicity. Spn-H 2 O 2 -induced lung toxicity causes the oxidation and non-proteolytic degradation of intracellular proteins tubulin, actin, and GAPDH. The ABC transporter GlnPQ is a heme-binding complex that makes Spn susceptible to heme toxicity.

4.
bioRxiv ; 2023 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-37333138

RESUMO

Streptococcus pneumoniae (Spn) causes pneumonia that kills millions through acute toxicity and invasion of the lung parenchyma. During aerobic respiration, Spn releases hydrogen peroxide (Spn-H 2 O 2 ), as a by-product of enzymes SpxB and LctO, and causes cell death with signs of both apoptosis and pyroptosis by oxidizing unknown cell targets. Hemoproteins are molecules essential for life and prone to oxidation by H 2 O 2 . We recently demonstrated that during infection-mimicking conditions, Spn-H 2 O 2 oxidizes the hemoprotein hemoglobin (Hb), releasing toxic heme. In this study, we investigated details of the molecular mechanism(s) by which the oxidation of hemoproteins by Spn-H 2 O 2 causes human lung cell death. Spn strains, but not H 2 O 2 -deficient SpnΔ spxB Δ lctO strains caused time-dependent cell cytotoxicity characterized by the rearrangement of the actin, the loss of the microtubule cytoskeleton and nuclear contraction. Disruption of the cell cytoskeleton correlated with the presence of invasive pneumococci and an increase of intracellular reactive oxygen species. In cell culture, the oxidation of Hb or cytochrome c (Cyt c ) caused DNA degradation and mitochondrial dysfunction from inhibition of complex I-driven respiration, which was cytotoxic to human alveolar cells. Oxidation of hemoproteins resulted in the creation of a radical, which was identified as a protein derived side chain tyrosyl radical by using electron paramagnetic resonance (EPR). Thus, we demonstrate that Spn invades lung cells, releasing H 2 O 2 that oxidizes hemoproteins, including Cyt c , catalyzing the formation of a tyrosyl side chain radical on Hb and causing mitochondrial disruption, that ultimately leads to the collapse of the cell cytoskeleton.

5.
bioRxiv ; 2023 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-38234816

RESUMO

Aim: Streptococcus pneumoniae (Spn) acquires genes for macrolide resistance, MEGA or ermB, in the human host. These genes are carried either in the chromosome, or on integrative conjugative elements (ICEs). Here, we investigated molecular determinants of the acquisition of macrolide resistance. Methods and Results: Whole genome analysis was conducted for 128 macrolide-resistant pneumococcal isolates to identify the presence of MEGA (44.5%, 57/128) or ermB (100%), and recombination events in Tn916-related elements or in the locus comCDE encoding competence genes. Confocal and electron microscopy studies demonstrated that, during the acquisition of macrolide resistance, pneumococcal strains formed clusters of varying size, with the largest aggregates having a median size of ~1600 µm2. Remarkably, these pneumococcal aggregates comprise both encapsulated and nonencapsulated pneumococci, exhibited physical interaction, and spanned extracellular and intracellular compartments. We assessed the recombination frequency (rF) for the acquisition of macrolide resistance by a recipient D39 strain, from pneumococcal strains carrying MEGA (~5.4 kb) in the chromone, or in large ICEs (>23 kb). Notably, the rF for the acquisition of MEGA, whether in the chromosome or carried on an ICE was similar. However, the rF adjusted to the acquisition of the full-length ICE (~52 kb), compared to that of the capsule locus (~23 kb) that is acquired by transformation, was three orders of magnitude higher. Finally, metabolomics studies revealed a link between the acquisition of ICE and the metabolic pathways involving nicotinic acid and sucrose. Conclusions: Extracellular and intracellular pneumococcal clusters facilitate the acquisition of full-length ICE at a rF higher than that of typical transformation events, involving distinct metabolic changes that present potential targets for interventions.

6.
Infect Immun ; 90(12): e0047122, 2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-36409115

RESUMO

Streptococcus pneumoniae (Spn) strains cause pneumonia that kills millions every year worldwide. Spn produces Ply, a hemolysin that lyses erythrocytes releasing hemoglobin, and also produces the pro-oxidant hydrogen peroxide (Spn-H2O2) during growth. The hallmark of the pathophysiology of hemolytic diseases is the oxidation of hemoglobin, but oxidative reactions catalyzed by Spn-H2O2 have been poorly studied. We characterized the oxidation of hemoglobin by Spn-H2O2. We prepared a series of single-mutant (ΔspxB or ΔlctO), double-mutant (ΔspxB ΔlctO), and complemented strains in TIGR4, D39, and EF3030. We then utilized an in vitro model with oxyhemoglobin to demonstrate that oxyhemoglobin was oxidized rapidly, within 30 min of incubation, by Spn-H2O2 to methemoglobin and that the main source of Spn-H2O2 was pyruvate oxidase (SpxB). Moreover, extended incubation caused the release and the degradation of heme. We then assessed oxidation of hemoglobin and heme degradation by other bacterial inhabitants of the respiratory tract. All hydrogen peroxide-producing streptococci tested caused the oxidation of hemoglobin and heme degradation, whereas bacterial species that produce <1 µM H2O2 neither oxidized hemoglobin nor degraded heme. An ex vivo bacteremia model confirmed that oxidation of hemoglobin and heme degradation occurred concurrently with hemoglobin that was released from erythrocytes by Ply. Finally, gene expression studies demonstrated that heme, but not red blood cells or hemoglobin, induced upregulated transcription of the spxB gene. Oxidation of hemoglobin may be important for pathogenesis and for the symbiosis of hydrogen peroxide-producing bacteria with other species by providing nutrients such as iron.


Assuntos
Heme , Peróxido de Hidrogênio , Peróxido de Hidrogênio/farmacologia , Heme/metabolismo , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo , Oxiemoglobinas/metabolismo , Hemoglobinas/metabolismo , Streptococcus/metabolismo , Oxirredução , Estresse Oxidativo , Catálise
7.
Microbiol Res ; 263: 127134, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35905580

RESUMO

Streptococcus pneumoniae (Spn) kills Staphylococcus aureus (Sau) through a contact-dependent mechanism that is catalyzed by cations, including iron, to convert hydrogen peroxide (H2O2) to highly toxic hydroxyl radicals (•OH). There are two well-characterized ABC transporters that contribute to the pool of iron in Spn, named Pia and Piu. Some Spn strains have acquired genes mef(E)/mel encoding another ABC trasporter (Mega) that produces an inducible efflux pump for resistance to macrolides. In macrolide-resistant Spn clinical isolates the insertion of Mega class 1. IV and 2. IVc deleted the locus piaABCD and these strains were attenuated for intoxicating Sau. The goal of this study was to investigate if the disruption of iron acquisition, or the antimicrobial-resistance activity of Mega, contributed to inhibiting the killing mechanism. Neither depletion of iron with 2,2'-dipyridyl-d8 (DP) nor incubating with a double knockout mutant SpnΔpiaAΔpiuA, inhibited killing of Sau. Clinical Spn strains carrying Mega1. IV or Mega2. IVc showed a significant delay for killing Sau. An ex vivo recombination system was used to transfer Mega1. IV or Mega2. IVc to reference Spn strains, which was confirmed by whole genome sequencing, and recombinants TIGR4Mega2. IVc, D39Mega2. IVc, and D39Mega1. IV were delayed for killing Sau. We then compared Sau killing of selected Mega-carrying Spn strains when incubated with sub-inhibitory erythromycin (Mega-induced) or sub-inhibitory cefuroxime. Remarkably, killing of Sau was completely inhibited under the Mega-induced condition whereas incubation with cefuroxime did not interfere with killing. Both mef(E) and mel were upregulated > 400-fold, and spxB (encoding an enzyme responsible for production of most H2O2) was upregulated 14.2-fold, whereas transcription of the autolysin (lytA) gene was downregulated when incubated with erythromycin. We demonstrated that erythromycin induction of Mega inhibits the •OH-mediated intoxication of Sau and that the inhibition occurred at the post-translational level suggesting that an imbalance of ions in the membrane inhibits these reactions.


Assuntos
Infecções Estafilocócicas , Streptococcus pneumoniae , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Cefuroxima , Farmacorresistência Bacteriana/genética , Eritromicina/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Ferro , Macrolídeos/farmacologia , Testes de Sensibilidade Microbiana , Staphylococcus aureus/genética , Streptococcus pneumoniae/genética
8.
Infect Immun ; 89(12): e0046321, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34543118

RESUMO

Streptococcus pneumoniae colonizes the nasopharynx of children and the elderly but also kills millions worldwide yearly. The secondary bile acid metabolite deoxycholic acid (DoC) affects the viability of human pathogens but also plays multiple roles in host physiology. We assessed in vitro the antimicrobial activity of DoC and investigated its potential to eradicate S. pneumoniae colonization using a model of human nasopharyngeal colonization and an in vivo mouse model of colonization. At a physiological concentration, DoC (0.5 mg/ml; 1.27 mM) killed all tested S. pneumoniae strains (n = 48) 2 h postinoculation. The model of nasopharyngeal colonization showed that DoC eradicated colonization by S. pneumoniae strains as soon as 10 min postexposure. The mechanism of action did not involve activation of autolysis, since the autolysis-defective double mutants ΔlytAΔlytC and ΔspxBΔlctO were as susceptible to DoC as was the wild type (WT). Oral streptococcal species (n = 20), however, were not susceptible to DoC (0.5 mg/ml). Unlike trimethoprim, whose spontaneous resistance frequency (srF) for TIGR4 or EF3030 was ≥1 × 10-9, no spontaneous resistance was observed with DoC (srF, ≥1 × 10-12). Finally, the efficacy of DoC to eradicate S. pneumoniae colonization was assessed in vivo using a topical route via intranasal (i.n.) administration and as a prophylactic treatment. Mice challenged with S. pneumoniae EF3030 carried a median of 4.05 × 105 CFU/ml 4 days postinoculation compared to 6.67 × 104 CFU/ml for mice treated with DoC. Mice in the prophylactic group had an ∼99% reduction of the pneumococcal density (median, 2.61 × 103 CFU/ml). Thus, DoC, an endogenous human bile salt, has therapeutic potential against S. pneumoniae.


Assuntos
Ácido Desoxicólico/farmacologia , Interações Hospedeiro-Patógeno , Infecções Pneumocócicas/microbiologia , Infecções Pneumocócicas/prevenção & controle , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/crescimento & desenvolvimento , Animais , Ácidos e Sais Biliares/metabolismo , Ácido Desoxicólico/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Farmacorresistência Bacteriana , Humanos , Camundongos , Mutação , N-Acetil-Muramil-L-Alanina Amidase/genética , Nasofaringe/microbiologia , Infecções Pneumocócicas/metabolismo , Streptococcus pneumoniae/genética
9.
Front Cell Infect Microbiol ; 11: 676638, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34295834

RESUMO

Introduction: Staphylococcus aureus strains, including methicillin-resistant S. aureus (MRSA) and methicillin-sensitive S. aureus (MSSA), are a main cause of nosocomial infection in the world. The majority of nosocomial S. aureus-infection are traced back to a source of contaminated surfaces including surgery tables. We assessed the efficacy of a mixture of levulinic acid (LA) and sodium dodecyl sulfate (SDS), hereafter called MoWa, to eradicate nosocomial pathogens from contaminated surfaces. Methods and Results: A dose response study demonstrated that MoWa killed 24 h planktonic cultures of S. aureus strains starting at a concentration of (LA) 8.2/(SDS) 0.3 mM while 24 h preformed biofilms were eradicated with 32/1.3 mM. A time course study further showed that attached MRSA bacteria were eradicated within 4 h of incubation with 65/2 mM MoWa. Staphylococci were killed as confirmed by bacterial counts, and fluorescence micrographs that were stained with the live/dead bacterial assay. We then simulated contamination of hospital surfaces by inoculating bacteria on a surface prone to contamination. Once dried, contaminated surfaces were sprayed with MoWa or mock-treated, and treated contaminated surfaces were swabbed and bacteria counted. While bacteria in the mock-treated samples grew at a density of ~104 cfu/cm2, those treated for ~1 min with MoWa (1.0/0.04 M) had been eradicated below limit of detection. A similar eradication efficacy was obtained when surfaces were contaminated with other nosocomial pathogens, such as Klebsiella pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii, or Staphylococcus epidermidis. Conclusions: MoWa kills planktonic and biofilms made by MRSA and MSSA strains and showed great efficacy to disinfect MRSA-, and MSSA-contaminated, surfaces and surfaces contaminated with other important nosocomial pathogens.


Assuntos
Infecção Hospitalar , Desinfetantes , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Desinfetantes/farmacologia , Hospitais , Humanos , Staphylococcus aureus
10.
J Ind Microbiol Biotechnol ; 43(9): 1237-47, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27342241

RESUMO

Staphylococcus aureus is a major food-borne pathogen due to the production of enterotoxin and is particularly prevalent in contaminated milk and dairy products. The lactic acid bacteria (LAB) are widely used as biocontrol agents in fermented foods which can inhibit pathogenic flora. In our work, we investigated the influence of three strains of LAB (Lactobacillus plantarum, Streptococcus thermophilus and Enterococcus durans) on the relative expression of three enterotoxin genes (sea, sec, sell) and eight virulence and/or regulatory genes (sarA, saeS, codY, srrA, rot, hld/RNAIII, agrA/RNAII, sigB) in two S. aureus strains (MW2 and Sa1612) in TSB and reduced-fat milk (1.5 %) at 30 °C over a 24-h period. The tested LAB and S. aureus strains proved to be mutually non-competitive or only slightly competitive during co-cultivation. In addition, under the above-mentioned conditions, differential gene expression between the S. aureus MW2 and Sa1612 strains was well documented. S. aureus growth was changed in mixed culture with LAB; however, its effect on the repression of sea and sec expression correlated with production of these virulence factors. In comparison, the presence of LAB strains generally inhibited the expression of sec, sell, sarA, seaS, agrA/RNAII and hld/RNAIII genes. The effect of LAB strains presence on the expression of sea, codY, srrA, rot and sigB genes was medium, time, LAB and S. aureus strain specific. SEA and SEC production was significantly reduced in milk compared to TSB in pure culture. After the 24-h cultivation, S. aureus MW2 and Sa1612 SEC production was 187 and 331 times lower in milk compared to TSB, respectively (0.07 and 0.39 ng/mL in milk, versus 13.1 and 129.2 ng/mL in TSB, respectively). At the same time S. aureus MW2 and Sa1612 SEA production was 77 and 68 times lower in milk compared to TSB, respectively (0.99 and 0.17 ng/mL in milk, versus 76.4 and 11.5 ng/mL in TSB, respectively). This study has revealed new insights into the interaction between S. aureus and LAB (L. plantarum, S. thermophilus, E. durans) on the level of the expression and/or production of S. aureus enterotoxins, regulatory and virulence genes in different media, including milk. This study provides data which may improve the quality of food production.


Assuntos
Leite , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Animais , Técnicas de Cocultura , Meios de Cultura , Enterococcus/fisiologia , Enterotoxinas/biossíntese , Enterotoxinas/genética , Microbiologia de Alimentos , Genes Reguladores , Lactobacillus plantarum/fisiologia , Interações Microbianas , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/patogenicidade , Streptococcus thermophilus/fisiologia , Transcriptoma , Fatores de Virulência/genética
11.
J Microbiol Methods ; 107: 197-204, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25447888

RESUMO

Staphylococcus aureus produces a wide variety of superantigenic activity Staphylococcal enterotoxins (SE) and they are a major cause of food poisoning. These superantigens are associated with mobile genetic elements such as plasmids, prophages and S. aureus pathogenicity islands (SaPI). The presence of well-known eight SaPI integrase and 13 enterotoxin genes (sea, seb, sec, sed, see, seg, seh, sei, sej, sel, sek, seq, and tst) in 93 S. aureus strains were investigated. All S. aureus isolates were characterized by pulsed-field gel electrophoresis (PFGE), and the genes were detected using five sets of multiplex PCR (mPCR). The most predominant toxin genes were sea (19%), seb (15%), sec (54%), sell (48%), selk (46%), selq (52%), seg (22%), and sei (19%). Analysis showed that many S. aureus isolates harbored multiple toxin genes. An mPCR-based assay was developed for the determination of all SaPI and their superantigen gene combinations. Twenty three isolates revealed the gene combination sec, sell and tst, typical of the SaPIbov1 and SaPIn1/m1 pathogenicity islands. Twelve isolates revealed the selk and selq gene combination consistent with SaPI3. Eight isolates exhibited the sec and sell genes without the tst gene typical of SaPImw2. We established a correlation between superantigenic toxin genotypes in S. aureus in terms of combinations of toxin gene-encoding SaPI. These results provide a rapid method for determining superantigenic toxin genotypes in S. aureus strains. A total of 24 PFGE patterns were generated. To our knowledge, this is a first study analyzing the correlation of all known SaPI and their enterotoxins in S. aureus using mPCR.


Assuntos
Ilhas Genômicas , Reação em Cadeia da Polimerase Multiplex/métodos , Staphylococcus aureus/genética , Superantígenos/genética , Animais , Análise por Conglomerados , Eletroforese em Gel de Campo Pulsado , Enterotoxinas/genética , Contaminação de Alimentos , Microbiologia de Alimentos , Humanos , Staphylococcus aureus/classificação
12.
Food Microbiol ; 44: 54-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25084645

RESUMO

Staphylococcal food poisoning is a global problem. The gene encoding enterotoxin C (sec) has been reported several times as the most frequent enterotoxin gene identified in food poisoning cases caused by contaminated milk. In this study, the expression of sec was examined during the growth of Staphylococcus aureus in milk compared to routinely used laboratory media. Additionally, expression of several regulatory genes (sarA, saeS, codY, srrA, rot, hld, agrA, sigB) and other five enterotoxin genes (sea, seg, seh, sek, sel) were observed. It has been well established for that S. aureus is able to grow in milk and we found significantly reduced expression of sec in milk compared to the laboratory medium (P < 0.05). Here, we report the first study providing a comprehensive view on the expression of enterotoxin genes and its regulation in milk. The milk environment dramatically changed the expression profiles of several enterotoxin genes although staphylococcal growth was not affected at all. The mechanism of the reduction may be explained by downregulation of the agr system, although other factors are expected to be involved. The constituent of milk causing the inhibitory effect remains unidentified.


Assuntos
Enterotoxinas/genética , Enterotoxinas/metabolismo , Leite/microbiologia , Staphylococcus aureus/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Regulação para Baixo , Expressão Gênica , Staphylococcus aureus/genética
13.
Mol Biol Rep ; 41(8): 5005-18, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24728610

RESUMO

Among the bacteria groups, most of them are known to be beneficial to human being whereas only a minority is being recognized as harmful. The pathogenicity of bacteria is due, in part, to their rapid adaptation in the presence of selective pressures exerted by the human host. In addition, through their genomes, bacteria are subject to mutations, various rearrangements or horizontal gene transfer among and/or within bacterial species. Bacteria's essential metabolic functions are generally encoding by the core genes. Apart of the core genes, there are several number of mobile genetic elements (MGE) acquired by horizontal gene transfer that might be beneficial under certain environmental conditions. These MGE namely bacteriophages, transposons, plasmids, and pathogenicity islands represent about 15% Staphylococcus aureus genomes. The acquisition of most of the MGE is made by horizontal genomic islands (GEI), recognized as discrete DNA segments between closely related strains, transfer. The GEI contributes to the wide spread of microorganisms with an important effect on their genome plasticity and evolution. The GEI are also involve in the antibiotics resistance and virulence genes dissemination. In this review, we summarize the mobile genetic elements of S. aureus.


Assuntos
Bacteriófagos/genética , Cromossomos Bacterianos/genética , Elementos de DNA Transponíveis/genética , Transferência Genética Horizontal/genética , Ilhas Genômicas/genética , Sequências Repetitivas Dispersas/genética , Staphylococcus aureus/genética , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA