Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Biochem Soc Trans ; 49(2): 617-627, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33704415

RESUMO

The human skin microbiota forms a key barrier against skin pathogens and is important in modulating immune responses. Recent studies identify lactobacilli as endogenous inhabitants of healthy skin, while inflammatory skin conditions are often associated with a disturbed skin microbiome. Consequently, lactobacilli-based probiotics are explored as a novel treatment of inflammatory skin conditions through their topical skin application. This review focuses on the potential beneficial role of lactobacilli (family Lactobacillaceae) in the skin habitat, where they can exert multifactorial local mechanisms of action against pathogens and inflammation. On one hand, lactobacilli have been shown to directly compete with skin pathogens through adhesion inhibition, production of antimicrobial metabolites, and by influencing pathogen metabolism. The competitive anti-pathogenic action of lactobacilli has already been described mechanistically for common different skin pathogens, such as Staphylococcus aureus, Cutibacterium acnes, and Candida albicans. On the other hand, lactobacilli also have an immunomodulatory capacity associated with a reduction in excessive skin inflammation. Their influence on the immune system is mediated by bacterial metabolites and cell wall-associated or excreted microbe-associated molecular patterns (MAMPs). In addition, lactobacilli can also enhance the skin barrier function, which is often disrupted as a result of infection or in inflammatory skin diseases. Some clinical trials have already translated these mechanistic insights into beneficial clinical outcomes, showing that topically applied lactobacilli can temporarily colonize the skin and promote skin health, but more and larger clinical trials are required to generate in vivo mechanistic insights and in-depth skin microbiome analysis.


Assuntos
Antibiose/imunologia , Candida albicans/imunologia , Inflamação/imunologia , Lactobacillus/imunologia , Pele/imunologia , Staphylococcus aureus/imunologia , Antibiose/fisiologia , Aderência Bacteriana/imunologia , Bacteriocinas/imunologia , Bacteriocinas/metabolismo , Candida albicans/fisiologia , Humanos , Sistema Imunitário/imunologia , Sistema Imunitário/microbiologia , Inflamação/microbiologia , Lactobacillus/metabolismo , Lactobacillus/fisiologia , Pele/microbiologia , Pele/patologia , Staphylococcus aureus/fisiologia
2.
FEBS Lett ; 594(23): 3920-3942, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33040342

RESUMO

Bacteria produce under certain stress conditions bacteriocins and microcins that display antibacterial activity against closely related species for survival. Bacteriocins and microcins exert their antibacterial activity by either disrupting the membrane or inhibiting essential intracellular processes of the bacterial target. To this end, they can lyse bacterial membranes and cause subsequent loss of their integrity or nutrients, or hijack membrane receptors for internalisation. Both bacteriocins and microcins are ribosomally synthesised and several are posttranslationally modified, whereas others are not. Such peptides are also toxic to the producer bacteria, which utilise immunity proteins or/and dedicated ATP-binding cassette (ABC) transporters to achieve self-immunity and peptide export. In this review, we discuss the structure and mechanism of self-protection that is conferred by these ABC transporters.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Bacteriocinas/metabolismo , Farmacorresistência Bacteriana , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/metabolismo , Bacteriocinas/imunologia , Humanos
3.
BMC Microbiol ; 20(1): 273, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32867691

RESUMO

BACKGROUND: Pectobacterium carotovorum subsp. carotovorum belongs to the Enterobacteriaceae family, which causes soft-rot disease in numerous plants worldwide resulting in significant economic losses. Results from our previous studies showed that the strain H-rif-8-6 produces low-molecular-weight bacteriocin (LMWB) Carocin S1. Interestingly, TH22-10, the caroS1K:Tn5 insertional mutant in H-rif-8-6, loses Carocin S1 producing ability, but still produces other LMWBs which the indicator strain SP33 can detect. The SP33 is one of the many strains that are sensitive toward the cytotoxic effects of Carocin S3K, but not Carocin S1. The result revealed that H-rif-8-6 is a multiple-bacteriocin producing strain. RESULTS: In this study, a 4.1-kb DNA fragment was isolated from the chromosomal DNA of Pcc strain, H-rif-8-6, by a DNA probe using the caroS1K gene as the template. DNA sequencing and analysis by GenBank revealed two complete open reading frames (ORFs), designated ORF1 and ORF2, which were identified within the sequence fragment. ORF1 and ORF2, similar to the identified carocin S2 genes, encode the killer (Carocin S3K) and the immunity (Carocin S3I) proteins, respectively, which were homologous to the colicin E3 gene. Carocin S3K and Carocin S3I were expressed, isolated, and purified in Escherichia coli BL21 after subcloning of the expression plasmid pGS3KI or pGSK3I. SDS-PAGE analysis showed that the relative masses of Carocin S3K and Carocin S3I were 95.6 kDa and 10.2 kDa, respectively. The results reveal that Carocin S3K has higher antimicrobial and specific antimicrobial activities for Pcc along with a nuclease activity than Carocin S3I. However, Carocin S3I inhibits the activity of Carocin S3K. Interestingly, a high concentration of Carocin S3I protein is also a DNA nuclease, and Carocin S3K also inhibits its activity. CONCLUSION: This study showed that another type of bacteriocin was found in Pectobacterium carotovorum. This new type of bacteriocin, Carocin S3, has the killer protein, Carocin S3K, and the immunity protein, Carocin S3I.


Assuntos
Bacteriocinas/genética , Bacteriocinas/farmacologia , Pectobacterium/genética , Bacteriocinas/química , Bacteriocinas/imunologia , Clonagem Molecular , Desoxirribonucleases/metabolismo , Escherichia coli/genética , Biblioteca Gênica , Peso Molecular , Pectobacterium/efeitos dos fármacos , Pectobacterium/metabolismo , Espectrometria de Massas por Ionização por Electrospray
4.
Genes (Basel) ; 11(7)2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32679707

RESUMO

Itaconic acid is an immunoregulatory metabolite produced by macrophages in response to pathogen invasion. It also exhibits antibacterial activity because it is an uncompetitive inhibitor of isocitrate lyase, whose activity is required for the glyoxylate shunt to be operational. Some bacteria, such as Yersinia pestis, encode enzymes that can degrade itaconic acid and therefore eliminate this metabolic inhibitor. Studies, primarily with Salmonella enterica subspecies enterica serovar Typhimurium, have demonstrated the presence of similar genes in this pathogen and the importance of these genes for the persistence of the pathogen in murine hosts. This minireview demonstrates that, based on Blast searches of 1063 complete Salmonella genome sequences, not all Salmonella serovars possess these genes. It is also shown that the growth of Salmonella isolates that do not possess these genes is sensitive to the acid under glucose-limiting conditions. Interestingly, most of the serovars without the three genes, including serovar Typhi, harbor DNA at the corresponding genomic location that encodes two open reading frames that are similar to bacteriocin immunity genes. It is hypothesized that these genes could be important for Salmonella that finds itself in strong competition with other Enterobacteriacea in the intestinal tract-for example, during inflammation.


Assuntos
Bacteriocinas/genética , Salmonella enterica/genética , Salmonella enterica/patogenicidade , Succinatos/metabolismo , Animais , Bacteriocinas/imunologia , Interações Hospedeiro-Patógeno/genética , Humanos , Intestinos/microbiologia , Isocitrato Liase/genética , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Salmonella enterica/enzimologia , Salmonella enterica/imunologia , Succinatos/imunologia , Yersinia pestis/enzimologia
5.
Nat Commun ; 10(1): 5288, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31754112

RESUMO

Bacterial pathogens often carry multiple prophages and other phage-derived elements within their genome, some of which can produce viral particles in response to stress. Listeria monocytogenes 10403S harbors two phage elements in its chromosome, both of which can trigger bacterial lysis under stress: an active prophage (ϕ10403S) that promotes the virulence of its host and can produce infective virions, and a locus encoding phage tail-like bacteriocins. Here, we show that the two phage elements are co-regulated, with the bacteriocin locus controlling the induction of the prophage and thus its activity as a virulence-associated molecular switch. More specifically, a metalloprotease encoded in the bacteriocin locus is upregulated in response to stress and acts as an anti-repressor for CI-like repressors encoded in each phage element. Our results provide molecular insight into the phenomenon of polylysogeny and its intricate adaptation to complex environments.


Assuntos
Bacteriófagos/imunologia , Cromossomos Bacterianos/imunologia , Listeria monocytogenes/imunologia , Prófagos/imunologia , Sequência de Aminoácidos , Bacteriocinas/genética , Bacteriocinas/imunologia , Bacteriólise/imunologia , Bacteriófagos/genética , Bacteriófagos/fisiologia , Cromossomos Bacterianos/genética , Cromossomos Bacterianos/virologia , Genoma Bacteriano/genética , Genoma Bacteriano/imunologia , Genoma Viral/genética , Genoma Viral/imunologia , Interações Hospedeiro-Patógeno/imunologia , Listeria monocytogenes/genética , Listeria monocytogenes/virologia , Lisogenia/genética , Lisogenia/imunologia , Metaloproteases/genética , Metaloproteases/imunologia , Prófagos/genética , Prófagos/fisiologia , Homologia de Sequência de Aminoácidos , Ativação Viral/genética , Ativação Viral/imunologia
6.
Semin Cell Dev Biol ; 88: 129-137, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29432952

RESUMO

The gastrointestinal tract is a complex environment in which the host immune system interacts with a diverse array of microorganisms, both symbiotic and pathogenic. As such, mobilizing a rapid and appropriate antimicrobial response depending on the nature of each stimulus is crucial for maintaining the balance between homeostasis and inflammation in the gut. Here we focus on the mechanisms by which intestinal antimicrobial peptides regulate microbial communities during dysbiosis and infection. We also discuss classes of bacterial peptides that contribute to reducing enteric pathogen outgrowth. This review aims to provide a comprehensive overview on the interplay of diverse antimicrobial responses with enteric pathogens and the gut microbiota.


Assuntos
Bacteriocinas/imunologia , Defensinas/imunologia , Disbiose/prevenção & controle , Trato Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Animais , Bacteriocinas/biossíntese , Bacteriocinas/farmacologia , Catelicidinas/biossíntese , Catelicidinas/imunologia , Catelicidinas/farmacologia , Defensinas/biossíntese , Defensinas/farmacologia , Disbiose/imunologia , Disbiose/microbiologia , Microbioma Gastrointestinal/imunologia , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Expressão Gênica/imunologia , Humanos , Imunidade nas Mucosas/efeitos dos fármacos , Inflamação , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Lipocalina-2/biossíntese , Lipocalina-2/imunologia , Lipocalina-2/farmacologia , Muramidase/biossíntese , Muramidase/imunologia , Muramidase/farmacologia , Simbiose/imunologia
7.
Antonie Van Leeuwenhoek ; 111(12): 2349-2360, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30006688

RESUMO

The structural gene that encodes thurincin H, a bacteriocin produced by Bacillus thuringiensis, is harboured in a genetic cluster (thnP, E, D, R, A1, A2, A3, B, T, I) that controls its synthesis, modification, secretion and autoimmunity. The specific genes in the cassette that confer immunity in B. thuringiensis to thurincin H are unknown. To identify these immunity determinants, we generated constructs that were used to transform a natural thurincin H-sensitive B. thuringiensis strain (i.e. Btk 404), and resistance or susceptibility to the bacteriocin in resultant recombinants was evaluated. When Btk 404/pHT3101-ThnARDEP and Btk 404/pHT3101-ThnABTI were exposed to thurincin H, immunity was demonstrated by the former only, indicating that ThnI does not play a role in resistance to the bacteriocin as previously proposed. Furthermore, we generated different sub-cassettes under the control of divergent promoters pThnR and pThur of the thurincin H locus, and pChi, and using the green fluorescent protein gene as reporter, which demonstrated that all promoters were recognised by ThnR, except pChi. We show for the first time that the small operon composed of thnR, thnD and thnE is required for immunity of B. thuringiensis to thurincin H, and thnI is not necessary for this response.


Assuntos
Transportadores de Cassetes de Ligação de ATP/imunologia , Bacillus thuringiensis/imunologia , Bacteriocinas/imunologia , Transportadores de Cassetes de Ligação de ATP/genética , Autoimunidade , Bacillus thuringiensis/efeitos dos fármacos , Bacillus thuringiensis/genética , Bacteriocinas/genética , Bacteriocinas/farmacologia , Regulação Bacteriana da Expressão Gênica , Genes Reguladores , Família Multigênica , Óperon , Regiões Promotoras Genéticas
8.
Curr Genet ; 64(2): 345-351, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28983718

RESUMO

This review attempts to analyze the mechanism of action and immunity of class IIa bacteriocins. These peptides are promising alternative food preservatives and they have a great potential application in medical sciences. Class IIa bacteriocins act on the cytoplasmic membrane of Gram-positive cells dissipating the transmembrane electrical potential by forming pores. However, their toxicity and immunity mechanism remains elusive. Here we discuss the role of the mannose phosphotransferase system (man-PTS) as the receptor for class IIa bacteriocins and the influence of the membrane composition on the activity of these antimicrobial peptides. A model that is consistent with experimental results obtained by different researchers involves the non-specific binding of the bacteriocin to the negatively charged membrane of target bacteria. This step would facilitate a specific binding to the receptor protein, altering its functionality and forming an independent pore in which the bacteriocin is inserted in the membrane. An immunity protein could specifically recognize and block the pore. Bacteriocins function in bacterial ecosystems and energetic costs associated with their production are also discussed. Theoretical models based on solid experimental evidence are vital to understand bacteriocins mechanism of action and to promote new technological developments.


Assuntos
Antibacterianos/química , Bacteriocinas/química , Imunidade/genética , Pediocinas/química , Antibacterianos/imunologia , Antibacterianos/uso terapêutico , Bacteriocinas/imunologia , Conservação de Alimentos , Humanos , Imunidade/efeitos dos fármacos , Modelos Teóricos , Pediocinas/imunologia , Peptídeos/química , Peptídeos/imunologia
9.
Mol Microbiol ; 105(6): 922-933, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28692133

RESUMO

The role of the class IIa bacteriocin membrane receptor protein remains unclear, and the following two different mechanisms have been proposed: the bacteriocin could interact with the receptor changing it to an open conformation or the receptor might act as an anchor allowing subsequent bacteriocin insertion and membrane disruption. Bacteriocin-producing cells synthesize an immunity protein that forms an inactive bacteriocin-receptor-immunity complex. To better understand the molecular mechanism of enterocin CRL35, the peptide was expressed as the suicidal probe EtpM-enterocin CRL35 in Escherichia coli, a naturally insensitive microorganism since it does not express the receptor. When the bacteriocin is anchored to the periplasmic face of the plasma membrane through the bitopic membrane protein, EtpM, E. coli cells depolarize and die. Moreover, co-expression of the immunity protein prevents the deleterious effect of EtpM-enterocin CRL35. The binding and anchoring of the bacteriocin to the membrane has demonstrated to be a sufficient condition for its membrane insertion. The final step of membrane disruption by EtpM-enterocin CRL35 is independent from the receptor, which means that the mannose PTS might not be involved in the pore structure. In addition, the immunity protein can protect even in the absence of the receptor.


Assuntos
Bacteriocinas/metabolismo , Escherichia coli/metabolismo , Antibacterianos/farmacologia , Bacteriocinas/imunologia , Membrana Celular/metabolismo , Escherichia coli/efeitos dos fármacos , Proteínas de Escherichia coli/metabolismo , Listeria , Potenciais da Membrana/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Peptídeos/metabolismo , Periplasma/metabolismo
10.
Probiotics Antimicrob Proteins ; 9(3): 345-354, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28063116

RESUMO

Bacteriocin TSU4 is a novel antimicrobial peptide isolated from Catla catla gut isolate Lactobacillus animalis TSU4. It has been reported for its potential antimicrobial activity against fish pathogens and food spoilage bacteria. In vivo safety evaluation is necessary to determine its immunogenicity, toxicity, and importance in real-life applications. The present study was designed to evaluate the immunogenicity, acute and sub-chronic toxicity of bacteriocin TSU4 in BALB/c mice to ensure its safety in industrial application. Male BALB/c mice were administered intraperitoneally for immunogenicity assessment, by oral gavage with 50, 100, and 200 mg/kg/body weight for acute test and 0.5 mg/kg/day dose of bacteriocin TSU4 for sub-chronic toxicity test. Neither mortality nor any infections were observed during experimental period. There was no major increase in antibody titer during the immunogenicity test, and no mortality was observed during acute or sub-chronic toxicity tests. The LD50 value of bacteriocin TSU4 was found to be higher than 200 ± 0.45 mg/kg. No significant change in the serum biochemical markers, histopathological analysis and visual observation in spleen sizes was observed. These findings revealed that bacteriocin TSU4 is a non-immunogenic, safe, non-toxic, and could be a potential candidate for industrial applications in food preservation and aquaculture industries.


Assuntos
Antibacterianos/imunologia , Antibacterianos/farmacologia , Bacteriocinas/imunologia , Bacteriocinas/farmacologia , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Biomarcadores/sangue , Creatinina/urina , Rim/efeitos dos fármacos , Rim/metabolismo , Lactobacillus/química , Dose Letal Mediana , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Testes de Toxicidade Aguda , Testes de Toxicidade Crônica , Ureia/urina
11.
Infect Immun ; 84(5): 1424-1437, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26902727

RESUMO

Members of the Burkholderia cepacia complex (Bcc) cause chronic opportunistic lung infections in people with cystic fibrosis (CF), resulting in a gradual lung function decline and, ultimately, patient death. The Bcc is a complex of 20 species and is rarely eradicated once a patient is colonized; therefore, vaccination may represent a better therapeutic option. We developed a new proteomics approach to identify bacterial proteins that are involved in the attachment of Bcc bacteria to lung epithelial cells. Fourteen proteins were reproducibly identified by two-dimensional gel electrophoresis from four Bcc strains representative of two Bcc species: Burkholderia cenocepacia, the most virulent, and B. multivorans, the most frequently acquired. Seven proteins were identified in both species, but only two were common to all four strains, linocin and OmpW. Both proteins were selected based on previously reported data on these proteins in other species. Escherichia coli strains expressing recombinant linocin and OmpW showed enhanced attachment (4.2- and 3.9-fold) to lung cells compared to the control, confirming that both proteins are involved in host cell attachment. Immunoproteomic analysis using serum from Bcc-colonized CF patients confirmed that both proteins elicit potent humoral responses in vivo Mice immunized with either recombinant linocin or OmpW were protected from B. cenocepacia and B. multivorans challenge. Both antigens induced potent antigen-specific antibody responses and stimulated strong cytokine responses. In conclusion, our approach identified adhesins that induced excellent protection against two Bcc species and are promising vaccine candidates for a multisubunit vaccine. Furthermore, this study highlights the potential of our proteomics approach to identify potent antigens against other difficult pathogens.


Assuntos
Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/metabolismo , Bacteriocinas/metabolismo , Infecções por Burkholderia/prevenção & controle , Complexo Burkholderia cepacia/fisiologia , Células Epiteliais/microbiologia , Adesinas Bacterianas/imunologia , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Bacteriocinas/imunologia , Infecções por Burkholderia/imunologia , Fibrose Cística/imunologia , Fibrose Cística/microbiologia , Modelos Animais de Doenças , Escherichia coli/genética , Escherichia coli/fisiologia , Feminino , Expressão Gênica , Humanos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Resultado do Tratamento
12.
Appl Biochem Biotechnol ; 177(4): 909-22, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26276444

RESUMO

Plantaricins, one of bacteriocin produced by Lactobacillus plantarum, are already known to have activities against several pathogenic bacterium. L. plantarum U10 isolated from "tempoyak," an Indonesian fermented food, produced one kind of plantaricin designated as plantaricin W (plnW). The plnW is suggested as a putative membrane location of protein and has similar conserved motif which is important as immunity to bacteriocin itself. Thus, due to study about this plantaricin, several constructs have been cloned and protein was analyzed in Lactococcus lactis. In this study, plnW gene was successfully cloned into vector NICE system pNZ8148 and created the transformant named L. lactis NZ3900 pNZ8148-WU10. PlnW protein was 25.3 kDa in size. The concentration of expressed protein was significantly increased by 10 ng/mL nisin induction. Furthermore, PlnW exhibited protease activity with value of 2.22 ± 0.05 U/mL and specific activity about 1.65 ± 0.03 U/mg protein with 50 ng/mL nisin induction. Immunity study showed that the PlnW had immunity activity especially against plantaricin and rendered L. lactis recombinant an immunity broadly to other bacteriocins such as pediocin, fermentcin, and acidocin.


Assuntos
Bacteriocinas/genética , Bacteriocinas/imunologia , Fermentação , Manipulação de Alimentos , Lactobacillus plantarum/metabolismo , Lactococcus lactis/genética , Sequência de Aminoácidos , Bacteriocinas/biossíntese , Bacteriocinas/química , Sequência de Bases , Clonagem Molecular , Expressão Gênica , Vetores Genéticos/genética , Dados de Sequência Molecular
13.
Biochimie ; 118: 344-55, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26092421

RESUMO

Archaeocins are ribosomally-synthesized antimicrobial peptides or proteins produced by archaea. Halocins and sulfolobicins are produced by archaea belonging to the order Halobacteriales (Euryarchaeota) and Sulfolobales (Crenarchaeota), respectively. These weapons contribute helping the producer to prosper in spite of the microbial warfare. Given the fact that many archaea thrive in various extreme environments, archaeocins are challenged with inhospitable and destructive environmental conditions. Their structural features and mechanisms of action, which could be original, mostly remain to be deciphered. This review summarizes the present knowledge on halocins and sulfolobicins, the two classes of archaeocins that have been evidenced until now, and brings light on aspects of emerging research such as their ecological role or potential applications. Other antimicrobial compounds produced by archaea are also considered.


Assuntos
Archaea/imunologia , Proteínas Arqueais/imunologia , Bacteriocinas/imunologia , Sequência de Aminoácidos , Archaea/metabolismo , Proteínas Arqueais/metabolismo , Bacteriocinas/metabolismo , Dados de Sequência Molecular
14.
Microbiology (Reading) ; 161(Pt 4): 683-700, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25406453

RESUMO

Bacteriocins are prokaryotic proteins or peptides with antimicrobial activity. Most of them exhibit a broad spectrum of activity, inhibiting micro-organisms belonging to different genera and species, including many bacterial pathogens which cause human, animal or plant infections. Therefore, these substances have potential biotechnological applications in either food preservation or prevention and control of bacterial infectious diseases. However, there is concern that continuous exposure of bacteria to bacteriocins may select cells resistant to them, as observed for conventional antimicrobials. Based on the models already investigated, bacteriocin resistance may be either innate or acquired and seems to be a complex phenomenon, arising at different frequencies (generally from 10(-9) to 10(-2)) and by different mechanisms, even amongst strains of the same bacterial species. In the present review, we discuss the prevalence, development and molecular mechanisms involved in resistance to bacteriocins produced by Gram-positive bacteria. These mechanisms generally involve changes in the bacterial cell envelope, which result in (i) reduction or loss of bacteriocin binding or insertion, (ii) bacteriocin sequestering, (iii) bacteriocin efflux pumping (export) and (iv) bacteriocin degradation, amongst others. Strategies that can be used to overcome this resistance are also addressed.


Assuntos
Anti-Infecciosos , Antibiose , Bacteriocinas/biossíntese , Bactérias Gram-Positivas/metabolismo , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/imunologia , Bacteriocinas/imunologia , Parede Celular , Loci Gênicos , Bactérias Gram-Positivas/genética , Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Imunidade Inata , Proteólise
15.
J Vis Exp ; (91): e51876, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-25350516

RESUMO

Streptococcus pneumoniae colonizes the highly diverse polymicrobial community of the nasopharynx where it must compete with resident organisms. We have shown that bacterially produced antimicrobial peptides (bacteriocins) dictate the outcome of these competitive interactions. All fully-sequenced pneumococcal strains harbor a bacteriocin-like peptide (blp) locus. The blp locus encodes for a range of diverse bacteriocins and all of the highly conserved components needed for their regulation, processing, and secretion. The diversity of the bacteriocins found in the bacteriocin immunity region (BIR) of the locus is a major contributor of pneumococcal competition. Along with the bacteriocins, immunity genes are found in the BIR and are needed to protect the producer cell from the effects of its own bacteriocin. The overlay assay is a quick method for examining a large number of strains for competitive interactions mediated by bacteriocins. The overlay assay also allows for the characterization of bacteriocin-specific immunity, and detection of secreted quorum sensing peptides. The assay is performed by pre-inoculating an agar plate with a strain to be tested for bacteriocin production followed by application of a soft agar overlay containing a strain to be tested for bacteriocin sensitivity. A zone of clearance surrounding the stab indicates that the overlay strain is sensitive to the bacteriocins produced by the pre-inoculated strain. If no zone of clearance is observed, either the overlay strain is immune to the bacteriocins being produced or the pre-inoculated strain does not produce bacteriocins. To determine if the blp locus is functional in a given strain, the overlay assay can be adapted to evaluate for peptide pheromone secretion by the pre-inoculated strain. In this case, a series of four lacZ-reporter strains with different pheromone specificity are used in the overlay.


Assuntos
Bacteriocinas/biossíntese , Streptococcus pneumoniae/metabolismo , Bacteriocinas/genética , Bacteriocinas/imunologia , Percepção de Quorum , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/imunologia
16.
Appl Environ Microbiol ; 80(20): 6303-15, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25085495

RESUMO

Subtilomycin was detected from the plant endophytic strain Bacillus subtilis BSn5 and was first reported from B. subtilis strain MMA7. In this study, a gene cluster that has been proposed to be related to subtilomycin biosynthesis was isolated from the BSn5 genome and was experimentally validated by gene inactivation and heterologous expression. Comparison of the subtilomycin gene cluster with other verified related lantibiotic gene clusters revealed a particular organization of the genes apnI and apnT downstream of apnAPBC, which may be involved in subtilomycin immunity. Through analysis of expression of the apnI and/or apnT genes in the subtilomycin-sensitive strain CU1065 and inactivation of apnI and apnT in the producer strain BSn5, we showed that the single gene apnI, encoding a putative transmembrane protein, was responsible for subtilomycin immunity. To our knowledge, evidence for lantibiotic immunity that is solely dependent on a transmembrane protein is quite rare. Further bioinformatic analysis revealed the abundant presence of ApnI-like proteins that may be responsible for lantibiotic immunity in Bacillus and Paenibacillus. We cloned the paeI gene, encoding one such ApnI-like protein, into CU1065 and showed that it confers resistance to paenibacillin. However, no cross-resistance was detected between ApnI and PaeI, even though subtilomycin and paenibacillin share similar structures, suggesting that the protection provided by ApnI/ApnI-like proteins involves a specific-sequence recognition mechanism. Peptide release/binding assays indicated that the recombinant B. subtilis expressing apnI interacted with subtilomycin. Thus, ApnI represents a novel model for lantibiotic immunity that appears to be common.


Assuntos
Bacillus subtilis/genética , Proteínas de Bactérias/imunologia , Bacteriocinas/genética , Bacteriocinas/imunologia , Bacteriocinas/farmacologia , Família Multigênica , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/imunologia , Bacillus subtilis/metabolismo , Proteínas de Bactérias/genética , Clonagem Molecular , Farmacorresistência Bacteriana/genética , Farmacorresistência Bacteriana/imunologia , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Testes de Sensibilidade Microbiana
17.
Probiotics Antimicrob Proteins ; 6(3-4): 177-85, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24880436

RESUMO

Emerging resistance to antiviral agents is a growing public health concern worldwide as it was reported for respiratory, sexually transmitted and enteric viruses. Therefore, there is a growing demand for new, unconventional antiviral agents which may serve as an alternative to the currently used drugs. Meanwhile, published literature continues shedding the light on the potency of lactic acid bacteria (LAB) and their bacteriocins as antiviral agents. Health-promoting LAB probiotics may exert their antiviral activity by (1) direct probiotic-virus interaction; (2) production of antiviral inhibitory metabolites; and/or (3) via stimulation of the immune system. The aim of this review was to highlight the antiviral activity of LAB and substances they produce with antiviral activity.


Assuntos
Antivirais/administração & dosagem , Bacteriocinas/imunologia , Lactobacillaceae/imunologia , Probióticos/administração & dosagem , Viroses/tratamento farmacológico , Animais , Humanos , Lactobacillaceae/genética , Viroses/imunologia
18.
Curr Microbiol ; 69(4): 423-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24838664

RESUMO

The immunity proteins of pediocin-like bacteriocins possess a positively charged region which is located at the C-terminus in all three subclasses. It has been suggested that this region may be involved in directing the immunity protein to the surface of the bacterial cell membrane. The aim of this study was to determine whether the positively charged residue lysine-46 (K46) around the hydrophobic pocket played a key role for immunity activity of subgroup A immunity protein PedB. At first, heterologous expression of the immune gene pedB from Lactobacillus plantarum BM-1 rendered the sensitive Lactobacillus plantarum WQ0815 resistant to bacteriocin BM-1. Then, using site-directed mutagenesis, the residue K46 was replaced by five different amino-acid residues, including arginine (R), aspartate (D), glutamate (E), glutamine (Q), and threonine (T). Western blot analysis confirmed that all mutated pedB genes were successfully expressed in the host L. plantarum WQ0815. Bacteriocin activity assays subsequently showed that any substitution of the K46 residue significantly reduced its immunity activity. Our present results indicated that the positively charged residue K46 located near the hydrophobic pocket was essential for the functionality of the immunity protein PedB.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Bacteriocinas/química , Lactobacillus plantarum/genética , Lisina/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Proteínas de Bactérias/imunologia , Bacteriocinas/genética , Bacteriocinas/imunologia , Lactobacillus plantarum/química , Lactobacillus plantarum/imunologia , Lisina/imunologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Homologia de Sequência de Aminoácidos
19.
PLoS One ; 9(4): e96222, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24781647

RESUMO

Streptococcus iniae is a Gram-positive bacterium and a severe pathogen to a wide range of economically important fish species. In addition, S. iniae is also a zoonotic pathogen and can cause serious infections in humans. In this study, we identified from a pathogenic S. iniae strain a putative bacteriocin, Sil, and examined its biological activity. Sil is composed of 101 amino acid residues and shares 35.6% overall sequence identity with the lactococcin 972 of Lactococcus lactis. Immunoblot analysis showed that Sil was secreted by S. iniae into the extracellular milieu. Purified recombinant Sil (rSil) exhibited a dose-dependent inhibitory effect on the growth of Bacillus subtilis but had no impact on the growths of other 16 Gram-positive bacteria and 10 Gram-negative bacteria representing 23 different bacterial species. Treatment of rSil by heating at 50°C abolished the activity of rSil. rSil bound to the surface of B. subtilis but induced no killing of the target cells. Cellular study revealed that rSil interacted with turbot (Scophthalmus maximus) head kidney monocytes and inhibited the innate immune response of the cells, which led to enhanced cellular infection of S. iniae. Antibody blocking of the extracellular Sil produced by S. iniae significantly attenuated the infectivity of S. iniae. Consistent with these in vitro observations, in vivo study showed that administration of turbot with rSil prior to S. iniae infection significantly increased bacterial dissemination and colonization in fish tissues. Taken together, these results indicate that Sil is a novel virulence-associated bacteriostatic and an immunoregulator that promotes S. iniae infection by impairing the immune defense of host fish.


Assuntos
Bacteriocinas/imunologia , Doenças dos Peixes/imunologia , Linguados/imunologia , Linguados/microbiologia , Infecções Estreptocócicas/veterinária , Streptococcus/patogenicidade , Fatores de Virulência/imunologia , Animais , Bacteriocinas/química , Células Cultivadas , Doenças dos Peixes/microbiologia , Imunidade Inata , Monócitos/imunologia , Monócitos/microbiologia , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Streptococcus/química , Streptococcus/imunologia , Fatores de Virulência/química
20.
J Dairy Sci ; 97(7): 4115-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24767890

RESUMO

Heterologous expression of bacteriocin genetic determinants (or operons) has long been a research interest for the functional analysis of genes involved in bacteriocin biosynthesis, regulation, modification, and immunity. Previously, construction of genomic libraries of the bacteriocin producer strains was usually required to identify new bacteriocin operons, a method that is tedious and time consuming. For the first time, we directly amplified an 8.14-kb bioinformatically identified thurincin H gene cluster using a one-step PCR method with 100% accuracy. This amplified gene cluster was cloned into plasmid pHT315, resulting in plasmid pGW139, and subsequently transformed to Bacillus thuringiensis EG10368, a strain naturally sensitive to thurincin H. Heterologous expression of the gene cluster makes the sensitive B. thuringiensis EG10368 produce thurincin H at a higher level compared with the wild-type producer, B. thuringiensis SF361. Moreover, B. thuringiensis EG10368pGW139 acquired complete immunity to thurincin H. The results indicated that one-step PCR is a promising tool to accurately amplify long bacteriocin gene clusters used in bacteriocin functional analysis studies and it is an effective way to produce bacteriocins at a higher level, without the need to clone large chromosomal fragments.


Assuntos
Bacillus thuringiensis/genética , Bacteriocinas/genética , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Família Multigênica , Bacillus thuringiensis/imunologia , Bacillus thuringiensis/metabolismo , Bacteriocinas/imunologia , Bacteriocinas/metabolismo , Laticínios/microbiologia , Conservantes de Alimentos/química , Amplificação de Genes , Plasmídeos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...