Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 226
Filtrar
1.
Methods Mol Biol ; 2839: 99-110, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39008250

RESUMO

Metal ion homeostasis in mitochondria is essential to maintaining proper cellular physiology. However, the ability of metals to bind off target or form complexes with multiple metabolites presents major challenges to understanding the mechanisms that govern this homeostasis. Adding further to the complexity, some of the major mitochondrial transporters have shown substrate promiscuity. In many cases, mitochondrial metals are found in the matrix compartment that is surrounded by the impermeable inner membrane. Four major classes of transporters facilitate the movement of solute across the inner membrane. These are mitochondrial carrier family, ATP-binding cassette transporters, mitochondrial pyruvate carriers, and sideroflexins. For iron, the matrix is the site of iron-sulfur clusters and heme synthesis and therefore transport must occur in a coordinated fashion with the cellular needs for these critical cofactors. Iron could be transported in numerous forms as it has been shown to form complexes with abundant metabolites such as citrate, nucleotides, or glutathione. Here, we describe assays to study iron (or any metal) transport by mitochondrial carrier family proteins expressed in Lactococcus lactis using a nisin-controlled expression system.


Assuntos
Ferro , Lactococcus lactis , Lactococcus lactis/metabolismo , Lactococcus lactis/genética , Ferro/metabolismo , Metais/metabolismo , Mitocôndrias/metabolismo , Transporte Biológico , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/genética , Nisina/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/genética
2.
Anal Chem ; 96(28): 11247-11254, 2024 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-38941069

RESUMO

Evaluating the dynamic interaction of microorganisms and mammalian cells is challenging due to the lack of suitable platforms for examining interspecies interactions in biologically relevant coculture conditions. In this work, we demonstrate the interaction between probiotic bacteria (Lactococcus lactis and Escherichia coli) and A498 human cancer cells in vitro, utilizing a hydrogel-based platform in a label-free manner by infrared spectroscopy. The L. lactis strain recapitulated in the compartment system secretes polypeptide molecules such as nisin, which has been reported to trigger cell apoptosis. We propose a mid-infrared (IR) spectroscopic imaging approach to monitor the variation of biological components utilizing kidney cells (A498) as a model system cocultured with bacteria. We characterized the biochemical composition (i.e., nucleic acids, protein secondary structures, and lipid conformations) label-free using an unbiased measurement. Several IR spectral features, including unsaturated fatty acids, ß-turns in protein, and nucleic acids, were utilized to predict cellular response. These features were then applied to establish a quantitative relationship through a multivariate regression model to predict cellular dynamics in the coculture system to assess the effect of nisin on A498 kidney cancer cells cocultured with bacteria. Overall, our study sheds light on the potential of using IR spectroscopic imaging as a label-free tool to monitor complex microbe-host cell interactions in biological systems. This integration will enable mechanistic studies of interspecies interactions with insights into their underlying physiological processes.


Assuntos
Técnicas de Cocultura , Escherichia coli , Probióticos , Humanos , Escherichia coli/metabolismo , Probióticos/metabolismo , Nisina/farmacologia , Nisina/química , Nisina/metabolismo , Lactococcus lactis/metabolismo , Espectrofotometria Infravermelho , Linhagem Celular Tumoral
3.
Sci Rep ; 14(1): 7292, 2024 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-38538691

RESUMO

Glioblastoma multiforme (GBM) IDH-wildtype is the most prevalent brain malignancy in adults. However, molecular mechanisms, which leads to GBM have not been completely elucidated. Granulocyte colony-stimulating factor (GCSF), Granulocyte colony-stimulating factor receptor GCSFR, and Signal transducers and activators of transcription 3 (STAT3) have been involved in the occurrence and development of various cancers, but their role in GBM is little known. Herein, we have investigated the gene and protein expression of GCSF, GCSFR, and STAT3 in 21 tissue biopsy samples and also in tumor associated normal tissue (TANT) samples derived from glioblastoma patients, which revealed significantly differential expression of these genes. To validate our findings, we performed a comprehensive integrated analysis of transcriptomic and proteomic profiling of respective genes by retrieving GBM RNA-sequence data from Genome Atlas Databases. GO and KEGG analysis revealed enrichment in disease-related pathways, such as JAK/STAT pathway activation, which were associated with GBM progression. We further performed computational docking analysis of potential drug candidate Nisin against GCSF, and the results were validated in vitro through cytotoxic activity assay using a human glioblastoma cell line SF-767 in a dose-dependent manner. Our comprehensive analysis reveals that GCSF augments glioma progression, and its blockade with anticancer bacteriocin peptide Nisin can potentially inhibit the growth and metastasis of GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Nisina , Adulto , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Nisina/metabolismo , Janus Quinases/metabolismo , Proteômica , Transdução de Sinais , Fatores de Transcrição STAT/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Fator Estimulador de Colônias de Granulócitos/metabolismo , Regulação Neoplásica da Expressão Gênica
4.
Nutrients ; 16(5)2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38474811

RESUMO

Lactic-acid-bacteria-derived bacteriocins are used as food biological preservatives widely. Little information is available on the impact of bacteriocin intake with food on gut microbiota in vivo. In this study, the effects of fermented milk supplemented with nisin (FM-nisin) or plantaricin Q7 (FM-Q7) from Lactiplantibacillus plantarum Q7 on inflammatory factors and the gut microbiota of mice were investigated. The results showed that FM-nisin or FM-Q7 up-regulated IFN-γ and down-regulated IL-17 and IL-12 in serum significantly. FM-nisin down-regulated TNF-α and IL-10 while FM-Q7 up-regulated them. The results of 16S rRNA gene sequence analysis suggested that the gut microbiome in mice was changed by FM-nisin or FM-Q7. The Firmicutes/Bacteroides ratio was reduced significantly in both groups. It was observed that the volume of Akkermansia_Muciniphila was significantly reduced whereas those of Lachnospiraceae and Ruminococcaceae were increased. The total number of short-chain fatty acids (SCFAs) in the mouse feces of the FM-nisin group and FM-Q7 group was increased. The content of acetic acid was increased while the butyric acid content was decreased significantly. These findings indicated that FM-nisin or FM-Q7 could stimulate the inflammation response and alter gut microbiota and metabolic components in mice. Further in-depth study is needed to determine the impact of FM-nisin or FM-Q7 on the host's health.


Assuntos
Microbioma Gastrointestinal , Lactobacillales , Nisina , Camundongos , Animais , Nisina/metabolismo , Nisina/farmacologia , Leite/metabolismo , RNA Ribossômico 16S/genética , Lactobacillales/metabolismo , Ácido Butírico
5.
NPJ Biofilms Microbiomes ; 10(1): 3, 2024 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-38233485

RESUMO

Oral microbiome dysbiosis mediates chronic periodontal disease, gut microbial dysbiosis, and mucosal barrier disfunction that leads to steatohepatitis via the enterohepatic circulation. Improving this dysbiosis towards health may improve liver disease. Treatment with antibiotics and probiotics have been used to modulate the microbial, immunological, and clinical landscape of periodontal disease with some success. The aim of the present investigation was to evaluate the potential for nisin, an antimicrobial peptide produced by Lactococcus lactis, to counteract the periodontitis-associated gut dysbiosis and to modulate the glycolipid-metabolism and inflammation in the liver. Periodontal pathogens, namely Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia and Fusobacterium nucleatum, were administrated topically onto the oral cavity to establish polymicrobial periodontal disease in mice. In the context of disease, nisin treatment significantly shifted the microbiome towards a new composition, commensurate with health while preventing the harmful inflammation in the small intestine concomitant with decreased villi structural integrity, and heightened hepatic exposure to bacteria and lipid and malondialdehyde accumulation in the liver. Validation with RNA Seq analyses, confirmed the significant infection-related alteration of several genes involved in mitochondrial dysregulation, oxidative phosphorylation, and metal/iron binding and their restitution following nisin treatment. In support of these in vivo findings indicating that periodontopathogens induce gastrointestinal and liver distant organ lesions, human autopsy specimens demonstrated a correlation between tooth loss and severity of liver disease. Nisin's ability to shift the gut and liver microbiome towards a new state commensurate with health while mitigating enteritis, represents a novel approach to treating NAFLD-steatohepatitis-associated periodontal disease.


Assuntos
Bacteriocinas , Nisina , Hepatopatia Gordurosa não Alcoólica , Doenças Periodontais , Camundongos , Humanos , Animais , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/metabolismo , Nisina/farmacologia , Nisina/metabolismo , Disbiose , Doenças Periodontais/microbiologia , Porphyromonas gingivalis/metabolismo , Inflamação/complicações , Estresse Oxidativo
6.
Prep Biochem Biotechnol ; 54(4): 494-502, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37607210

RESUMO

The production of Nisin, an FDA-approved food preservative, was attempted by Lactococcus lactis subsp. lactis ATCC® 11454 using the underutilized milk industry effluent, acid-whey, as a substrate. Nisin production was further improved by studying the effect of supplementation of nutrients and non-nutritional parameters. The addition of yeast extract (6% w/v) as nitrogen source and sucrose (4% w/v) as carbon source were found to be suitable nutrients for the maximum nisin production. The changes in the medium pH due to lactic acid accumulation during batch fermentation and its influence on the production of nisin were analyzed in the optimized whey medium (OWM). The production characteristics in OWM were further compared with the nisin production in MRS media. The influence of nisin as an inducer for its own production was also studied and found that the addition of nisin at 0.22 mg/ml promote the nisin production. The analysis of consumption of various metal ions present in the OWM during the nisin production was also analyzed, and found that the copper ions are the most consumed ion. The highest nisin yield of 2.6 × 105 AU/mL was obtained with OWM.


Assuntos
Lactococcus lactis , Nisina , Nisina/metabolismo , Soro do Leite/metabolismo , Lactococcus lactis/metabolismo , Proteínas do Soro do Leite , Fermentação , Suplementos Nutricionais , Íons , Meios de Cultura
7.
J Neuroinflammation ; 20(1): 228, 2023 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-37803465

RESUMO

INTRODUCTION: Periodontitis-related oral microbial dysbiosis is thought to contribute to Alzheimer's disease (AD) neuroinflammation and brain amyloid production. Since probiotics can modulate periodontitis/oral dysbiosis, this study examined the effects of a probiotic/lantibiotic, nisin, in modulating brain pathology triggered by periodontitis. METHODS: A polymicrobial mouse model of periodontal disease was used to evaluate the effects of this disease on brain microbiome dysbiosis, neuroinflammation, Alzheimer's-related changes, and nisin's therapeutic potential in this context. RESULTS: 16S sequencing and real-time PCR data revealed that Nisin treatment mitigated the changes in the brain microbiome composition, diversity, and community structure, and reduced the levels of periodontal pathogen DNA in the brain induced by periodontal disease. Nisin treatment significantly decreased the mRNA expression of pro-inflammatory cytokines (Interleukin-1ß/IL-1 ß, Interleukin 6/IL-6, and Tumor Necrosis Factor α/TNF-α) in the brain that were elevated by periodontal infection. In addition, the concentrations of amyloid-ß 42 (Aß42), total Tau, and Tau (pS199) (445.69 ± 120.03, 1420.85 ± 331.40, 137.20 ± 36.01) were significantly higher in the infection group compared to the control group (193.01 ± 31.82, 384.27 ± 363.93, 6.09 ± 10.85), respectively. Nisin treatment markedly reduced the Aß42 (261.80 ± 52.50), total Tau (865.37 ± 304.93), and phosphorylated Tau (82.53 ± 15.77) deposition in the brain of the infection group. DISCUSSION: Nisin abrogation of brain microbiome dysbiosis induces beneficial effects on AD-like pathogenic changes and neuroinflammation, and thereby may serve as a potential therapeutic for periodontal-dysbiosis-related AD.


Assuntos
Doença de Alzheimer , Bacteriocinas , Microbiota , Nisina , Periodontite , Probióticos , Camundongos , Animais , Doença de Alzheimer/patologia , Nisina/metabolismo , Bacteriocinas/metabolismo , Doenças Neuroinflamatórias , Disbiose/tratamento farmacológico , Disbiose/metabolismo , Periodontite/metabolismo , Encéfalo/metabolismo , Peptídeos beta-Amiloides/metabolismo , Interleucina-6/metabolismo , Probióticos/uso terapêutico
8.
FEMS Microbiol Rev ; 47(3)2023 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-37300874

RESUMO

It is almost a century since nisin was discovered in fermented milk cultures, coincidentally in the same year that penicillin was first described. Over the last 100 years this small, highly modified pentacyclic peptide has not only found success in the food industry as a preservative but has also served as the paradigm for our understanding of the genetic organization, expression, and regulation of genes involved in lantibiotic biosynthesis-one of the few cases of extensive post-translation modification in prokaryotes. Recent developments in understanding the complex biosynthesis of nisin have shed light on the cellular location of the modification and transport machinery and the co-ordinated series of spatio-temporal events required to produce active nisin and provide resistance and immunity. The continued unearthing of new natural variants from within human and animal gastrointestinal tracts has sparked interest in the potential application of nisin to influence the microbiome, given the growing recognition of the role the gastrointestinal microbiota plays in health and disease. Moreover, interdisciplinary approaches have taken advantage of biotechnological advancements to bioengineer nisin to produce novel variants and expand nisin functionality for applications in the biomedical field. This review will discuss the latest progress in these aspects of nisin research.


Assuntos
Bacteriocinas , Lactococcus lactis , Nisina , Humanos , Nisina/genética , Nisina/metabolismo , Bacteriocinas/metabolismo , Processamento de Proteína Pós-Traducional , Penicilinas/metabolismo , Antibacterianos/metabolismo , Lactococcus lactis/genética , Lactococcus lactis/metabolismo
9.
Bioresour Technol ; 385: 129387, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37364650

RESUMO

Glycosyltransferases synthesize a variety of exopolysaccharides (EPS) with different properties by altering the type of glycosidic linkage, degree of branching, length, mass, and conformation of the polymers. The genome analysis of an EPS-producing, Lactobacillus plantarum BR2 (Accession No: MN176402) showed twelve glycosyltransferase genes, and the gene BR2gtf (1116 bp), annotated as an EPS biosynthetic glycosyltransferase was cloned into the pNZ8148 vector. The recombinant pNZ8148 vector along with pNZ9530, a regulatory plasmid, were electroporated to L. plantarum BR2 for the over-expression of gtf gene under a nisin-controlled expression system and the glycosyltransferase activity of the recombinant and the wild-type strains were analysed. The recombinant strain showed 54.4% increase in EPS production with the maximum EPS production of 23.2 ± 0.5 g/L in a 5 L bioreactor study after 72 h of fermentation. This study shows an effective molecular strategy possibly to be adopted in lactic acid bacteria to enhance exopolysaccharide production.


Assuntos
Lactobacillales , Lactobacillus plantarum , Nisina , Lactobacillus plantarum/genética , Lactobacillus plantarum/metabolismo , Nisina/genética , Nisina/metabolismo , Lactobacillales/metabolismo , Plasmídeos , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Polissacarídeos Bacterianos/metabolismo
10.
Sci Rep ; 13(1): 7899, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37193715

RESUMO

Nisin is a broad spectrum bacteriocin used extensively as a food preservative that was identified in Lactococcus lactis nearly a century ago. We show that orally-ingested nisin survives transit through the porcine gastrointestinal tract intact (as evidenced by activity and molecular weight determination) where it impacts both the composition and functioning of the microbiota. Specifically, nisin treatment caused a reversible decrease in Gram positive bacteria, resulting in a reshaping of the Firmicutes and a corresponding relative increase in Gram negative Proteobacteria. These changes were mirrored by the modification in relative abundance of pathways involved in acetate, butyrate (decreased) and propionate (increased) synthesis which correlated with overall reductions in short chain fatty acid levels in stool. These reversible changes that occur as a result of nisin ingestion demonstrate the potential of bacteriocins like nisin to shape mammalian microbiomes and impact on the functionality of the community.


Assuntos
Bacteriocinas , Microbioma Gastrointestinal , Lactococcus lactis , Nisina , Animais , Suínos , Nisina/farmacologia , Nisina/metabolismo , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Bacteriocinas/farmacologia , Bacteriocinas/metabolismo , Bactérias Gram-Positivas/metabolismo , Lactococcus lactis/metabolismo , Mamíferos/metabolismo
11.
J Biomed Nanotechnol ; 18(4): 1227-1235, 2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35854440

RESUMO

Bacteriocins are peptides produced by bacteria to inhibit the growth of other prokaryotes. Nisin is a bacteriocin widely used in the food industry and for biomedical applications. However, bacteriocins have some limitations, as they experience mechanisms of resistance, degradation by proteases, and suboptimal intracellular delivery. Combining bacteriocins with nanoscale drug delivery systems (nano-DDS) is an approach that can help overcome these limitations. Among the nano-DDS, solid lipid nanoparticles (SLN) have been described as promising candidates, because of their potential for industrial scale-up and lower toxicity. The objective of this proof-of-concept study was to investigate the use of nisin-loaded SLN (SLN-Nisin) as an antimicrobial and anticancer therapeutic. We show that SLN-Nisin can significantly inhibit the growth of the oral pathogen, Treponema denticola, disrupt oral biofilms, and decrease oral squamous cell carcinoma cell (OSCC) viability compared to free nisin. Further, analysis with scanning electron microscopy (SEM) revealed significant morphological changes in OSCC cells challenged with SLN-Nisin, compared to the empty-nanoparticle or free nisin, indicating that SLN-Nisin likely decreases cell viability by increasing pore formation. This data reveals that nano-DDS are robust tools that can enhance bacteriocin properties.


Assuntos
Antineoplásicos , Bacteriocinas , Carcinoma de Células Escamosas , Neoplasias Bucais , Nanopartículas , Nisina , Antibacterianos/química , Antineoplásicos/farmacologia , Bacteriocinas/química , Bacteriocinas/metabolismo , Bacteriocinas/farmacologia , Biofilmes , Humanos , Lipossomos , Nisina/química , Nisina/metabolismo , Nisina/farmacologia
12.
Sci Rep ; 12(1): 4232, 2022 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35273305

RESUMO

Treatment of bacterial infections is one of the major challenges of our time due to the evolved resistance mechanisms of pathogens against antibiotics. To circumvent this problem, it is necessary to understand the mode of action of the drug and the mechanism of resistance of the pathogen. One of the most potent antibiotic targets is peptidoglycan (PGN) biosynthesis, as this is an exclusively occurring and critical feature of bacteria. Lipid II is an essential PGN precursor synthesized in the cytosol and flipped into the outer leaflet of the membrane prior to its incorporation into nascent PGN. Antimicrobial peptides (AMPs), such as nisin and colistin, targeting PGN synthesis are considered promising weapons against multidrug-resistant bacteria. However, human pathogenic bacteria that were also resistant to these compounds evolved by the expression of an ATP-binding cassette transporter of the bacitracin efflux (BceAB) type localized in the membrane. In the human pathogen Streptococcus agalactiae, the BceAB transporter SaNsrFP is known to confer resistance to the antimicrobial peptide nisin. The exact mechanism of action for SaNsrFP is poorly understood. For a detailed characterization of the resistance mechanism, we heterologously expressed SaNsrFP in Lactococcus lactis. We demonstrated that SaNsrFP conferred resistance not only to nisin but also to a structurally diverse group of antimicrobial PGN-targeting compounds such as ramoplanin, lysobactin, or bacitracin/(Zn)-bacitracin. Growth experiments revealed that SaNsrFP-producing cells exhibited normal behavior when treated with nisin and/or bacitracin, in contrast to the nonproducing cells, for which growth was significantly reduced. We further detected the accumulation of PGN precursors in the cytoplasm after treating the cells with bacitracin. This did not appear when SaNsrFP was produced. Whole-cell proteomic protein experiments verified that the presence of SaNsrFP in L. lactis resulted in higher production of several proteins associated with cell wall modification. These included, for example, the N-acetylmuramic acid-6-phosphate etherase MurQ and UDP-glucose 4-epimerase. Analysis of components of the cell wall of SaNsrFP-producing cells implied that the transporter is involved in cell wall modification. Since we used an ATP-deficient mutant of the transporter as a comparison, we can show that SaNsrFP and its inactive mutant do not show the same phenotype, albeit expressed at similar levels, which demonstrates the ATP dependency of the mediated resistance processes. Taken together, our data agree to a target protection mechanism and imply a direct involvement of SaNsrFP in resistance by shielding the membrane-localized target of these antimicrobial peptides, resulting in modification of the cell wall.


Assuntos
Nisina , Trifosfato de Adenosina , Antibacterianos/farmacologia , Bacitracina/farmacologia , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana/genética , Proteínas de Membrana Transportadoras/genética , Nisina/metabolismo , Nisina/farmacologia , Proteômica
13.
Bioresour Technol ; 348: 126776, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35104649

RESUMO

Nisin has a tendency to associate with the cell wall of the producing strain, which inhibits growth and lowers the ceiling for nisin production. With the premise that resistance to the cationic chlorhexidine could reduce nisin binding, variants with higher tolerance to this compound were isolated. One of the resistant isolates, AT0606, had doubled its resistance to nisin, and produced three times more free nisin, when cultured in shake flasks. Characterization revealed that AT0606 had an overall less negatively charged and thicker cell wall, and these changes appeared to be linked to a defect high-affinity phosphate uptake system, and a mutation inactivating the oleate hydratase. Subsequently, the potential of using AT0606 for cost efficient production of nisin was explored, and it was possible to attain a high titer of 13181 IU/mL using a fermentation substrate based on molasses and a by-product from whey protein hydrolysate production.


Assuntos
Lactococcus lactis , Nisina , Clorexidina/metabolismo , Fermentação , Lactococcus lactis/genética , Nisina/metabolismo , Nisina/farmacologia , Rios
14.
Biochem Cell Biol ; 100(2): 136-141, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34986025

RESUMO

Nisin, an antimicrobial peptide produced by Lactococcus lactis, is widely used as a safe food preservative and has recently attracted the attention of researchers as a potential anticancer agent. The cytotoxicity of nisin against human cervical cancer cell lines (HeLa), human ovarian carcinoma cell lines (OVCAR-3 and SK-OV-3), and human umbilical vein endothelial cells (HUVECs) was evaluated using an MTT assay. The apoptotic effect of nisin was identified by Annexin-V/propidium iodide assay, which was further confirmed by western blotting analysis, mitochondrial membrane potential (ΔΨm) analysis, and reactive oxygen species (ROS) assay. The MTT assay showed concentration-dependent cytotoxicity of nisin towards cancer cell lines, with IC50 values of 11.5-23 µM, but less toxicity against normal endothelial cells. Furthermore, the treatment of cervical cancer cells with 12 µM nisin significantly (P < 0.05) increased the Bax/Bcl-2 ratio (4.9 fold), reduced ΔΨm (70%), and elevated ROS levels (1.7 fold). These findings indicate that nisin may have anticancer and apoptogenic activities through mitochondrial dysfunction and oxidative stress damage in cervical cancer cells.


Assuntos
Nisina , Neoplasias Ovarianas , Neoplasias do Colo do Útero , Apoptose , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Feminino , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Nisina/metabolismo , Nisina/farmacologia , Neoplasias Ovarianas/patologia , Espécies Reativas de Oxigênio/metabolismo , Neoplasias do Colo do Útero/metabolismo
15.
Appl Environ Microbiol ; 88(2): e0161821, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-34788067

RESUMO

Lanthipeptides belong to a family of ribosomally synthesized and posttranslationally modified peptides (RiPPs) containing (methyl)lanthionine residues. Commonly, class I lanthipeptides are synthesized by a gene cluster encoding a precursor peptide (LanA), biosynthetic machinery (LanBTC), a protease (LanP), a two-component regulatory system (LanRK), and an immunity system (LanI and LanFEG). Although nisin and subtilin are highly similar class I lanthipeptides, the cross-regulation by LanRK and the cross-immunity by LanI and LanFEG between the nisin and subtilin systems have been proven to be very low. Here, the possibility of the cross-functionality of LanBTC to modify and transport nisin precursor (NisA) and subtilin precursor (SpaS) was evaluated in Bacillus subtilis and Lactococcus lactis. Interestingly, we found that a promiscuous NisBC-SpaT complex is able to synthesize and export nisin precursor, as efficiently as the native nisin biosynthetic machinery NisBTC, in L. lactis but not B. subtilis. The assembly of the NisBC-SpaT complex at a single microdomain, close to the old cell pole, was observed by fluorescence microscopy in L. lactis. In contrast, such a complex was not formed in B. subtilis. Furthermore, the isolation of the NisBC-SpaT complex and its subcomplexes from the cytoplasmic membrane of L. lactis by pulldown assays was successfully conducted. Our work demonstrates that the association of LanBC with LanT is critical for the efficient biosynthesis and secretion of the lanthipeptide precursor with complete modifications and suggests a cooperative mechanism between LanBC and LanT in the modification and transport processes. IMPORTANCE A multimeric synthetase LanBTC complex has been proposed for the in vivo production of class I lanthipeptides. However, it has been demonstrated that LanB, LanC, and LanT can perform their functionality in vivo and in vitro, independently of other Lan proteins. The role of protein-protein interactions, especially between the modification complex LanBC and the transport system LanT, in the biosynthesis process of lanthipeptides is still unclear. In this study, the importance of the presence of a well-installed LanBTC complex in the cell membrane for lanthipeptide biosynthesis and transport was reinforced. In L. lactis, the recruitment of SpaT from the peripheral cell membrane to the cell poles by the NisBC complex was observed, which may explain the mechanism by which the secretion of the premature peptide is prevented.


Assuntos
Lactococcus lactis , Nisina , Bacillus subtilis/genética , Proteínas de Bactérias/metabolismo , Transporte Biológico , Lactococcus lactis/metabolismo , Ligases/metabolismo , Nisina/metabolismo
16.
mBio ; 12(5): e0258521, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34607454

RESUMO

Nisin is synthesized by a putative membrane-associated lantibiotic synthetase complex composed of the dehydratase NisB, the cyclase NisC, and the ABC transporter NisT in Lactococcus lactis. Earlier work has demonstrated that NisB and NisT are linked via NisC to form such a complex. Here, we conducted for the first time the isolation of the intact NisBTC complex and NisT-associated subcomplexes from the cytoplasmic membrane by affinity purification. A specific interaction of NisT, not only with NisC but also with NisB, was detected. The cellular presence of NisB and/or NisC in complex with precursor nisin (NisA) was determined, which shows a highly dynamic and transient assembly of the NisABC complex via an alternating binding mechanism during nisin dehydration and cyclization. Mutational analyses, with cysteine-to-alanine mutations in NisA, suggest a tendency for NisA to lose affinity to NisC concomitant with an increasing number of completed lanthionine rings. Split NisBs were able to catalyze glutamylation and elimination reactions in an alternating way as efficiently as full-length NisB, with no significant influence on the following cyclization and transport. Notably, the harvest of the leader peptide in complex with the independent elimination domain of NisB points to a second leader peptide binding motif that is located in the C-terminal region of NisB, giving rise to a model where the leader peptide binds to different sites in NisB for glutamylation and elimination. Overall, these combined studies provide new insights into the cooperative biosynthesis mechanism of nisin and thereby lay a foundation for further structural and functional characterization of the NisBTC complex. IMPORTANCE Lantibiotics are ribosomally synthesized and posttranslationally modified peptide antibiotics. Although the membrane-associated lantibiotic biosynthesis machinery has long been proposed to exist, the isolation of such a complex has not been reported yet, which limits the elucidation of the processive mechanism of lantibiotic biosynthesis. In this work, we present direct evidence for the existence of the nisin biosynthetic complex at the cytoplasmic membrane of L. lactis, producing fully modified precursor nisin. By analyses of the interactions within the intact NisBTC complex and the modification machinery NisABC, we were able to elucidate the cooperative action for the modification and transport of nisin. Inspired by the natural and documented degradation process of NisB, artificial split-NisBs were made and thoroughly characterized, demonstrating a crucial clue to the evolution of the LanB family. Importantly, our study also suggests that the leader peptide of NisA binds to two different recognition motifs, i.e., one for glutamylation and one for elimination.


Assuntos
Proteínas de Bactérias/genética , Lactococcus lactis/genética , Nisina/genética , Nisina/metabolismo , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/metabolismo , Lactococcus lactis/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Nisina/biossíntese , Nisina/classificação , Processamento de Proteína Pós-Traducional , Sinais Direcionadores de Proteínas/genética
17.
J Biosci Bioeng ; 132(6): 606-612, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34563462

RESUMO

The transition of the bacterial biota of Kishu saba-narezushi (mackerel-narezushi) in the Hidaka region of Wakayama prefecture, Japan, was analyzed using amplicon sequencing based on the V3-V4 variable region of the 16S rRNA gene. In the non-fermented sample (0 day), the major genus with the highest abundance ratio was Staphylococcus. In the early stage (fermentation for 2 days), however, the genus Lactococcus became a dominant species, and in the later stage (fermentation for 5 days), the abundance ratio of the genus Lactobacillus increased significantly. Lactococcus lactis strains isolated from the narezushi samples had the ability to suppress the growth of not only Staphylococcus genera but also Lactobacillus. Moreover, the isolates produced a bacteriocin, which was identified as nisin Z. On the basis of these results, it is concluded that L. lactis plays an important role in preparing the fermentation conditions of Kishu saba-narezushi in the early stage by suppressing unwanted microorganisms using lactic acid and nisin Z.


Assuntos
Lactococcus lactis , Nisina , Perciformes , Animais , Bactérias/metabolismo , Biota , Fermentação , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Nisina/metabolismo , Perciformes/metabolismo , RNA Ribossômico 16S/genética
18.
Curr Microbiol ; 78(9): 3430-3438, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34255153

RESUMO

An attempt was made, to characterize natural antibiotics or lantibiotics from unconventional sources and its antibacterial spectrum against food borne pathogens and drug resistant bacteria. Six different traditional fermented foods i.e., fermented fish, fermented soybeans, Soibum (fermented bamboo shoots), milk, idly and dosa batter were used for the isolation of bacteriocin producing Lactic acid bacteria (LAB). Among all bacterial cultures isolated from the various sources, 129 cultures have found to produce antimicrobial compounds. Nisin specific reporter bacteria was utilized as biosensor to identify the Nisin like bacteriocin, where 10 cultures found to be positive Nisin producer. Identified Nisin like bacteriocin was partially concentrated by using ammonium sulphate followed by butanol extraction. Minimum inhibitory concentration (MIC) was analyzed against food borne pathogen and drug resistant bacteria. MIC of partially purified Nisin (pp-Nisin) of all the LAB isolates against food-borne pathogens are ranged between 0.5 and 92 µg/ml respected to various Gram-positive bacteria. Similarly, the drug resistant bacteria were also inhibited by pp-Nisin (MIC ranged between 15 and 175 µg/ml). All samples of ppnisin exhibited auto induction ability. Taxonomic identification of the nisin producers was done by whole genome sequencing which reveals that cultures belongs to Lactococcus lactis ssp. lactis. Also it was found that Lactococcus lactis ssp. lactis C2d and Lactococcus lactis ssp. lactis SP2C4 harbor nisA gene and Lactococcus lactis ssp. lactis FS2 (L. lactis FS2) harbor nisQ gene. The finding of this study highlights the first case of L. lactis FS2 isolated from fermented fish harbor nisQ gene. Antibacterial activity of pp-Nisin against drug resistant LAB is also reported.


Assuntos
Bacteriocinas , Alimentos Fermentados , Lactobacillales , Lactococcus lactis , Nisina , Animais , Bacteriocinas/genética , Fermentação , Lactobacillales/genética , Lactobacillales/metabolismo , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Nisina/metabolismo
19.
Food Microbiol ; 99: 103835, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34119119

RESUMO

In this study, we examined the ability of nisin A and a rationally assembled bank of 36 nisin derivative producing Lactococcus lactis strains to inhibit Listeria. A broth-based bioluminescence assay for screening single and combinations of bioengineered nisin derivatives using cell-free supernatants (CFS) from nisin derivative producing strains was developed. In this way, we screened 630 combinations of nisin derivative producing strains, identifying two (CFS from M17Q + N20P and M17Q + S29E) which exhibited enhanced anti-listerial activity when used together compared to when used alone, or to the nisin A producing strain. Minimal inhibitory concentration assays performed with purified peptides revealed than when used singly, the specific activities of M17Q, N20P and S29E (3.75-7.5 µM) against L. innocua were equal to, or less than that of nisin A (MIC of 3.75 µM). Broth-based growth curve assays using purified peptides demonstrated that use of the double peptide combinations and a triple peptide combination (M17Q + N20P + S29E) resulted in an extended lag phase of L. innocua, while kill curve assays confirmed the enhanced bactericidal activity of the combinations in comparison to the single derivative peptides or nisin A. Furthermore, the enhanced activity of the M17Q + N20P combination was maintained in a model food system (frankfurter homogenate) at both chill (4 °C) and abusive (20 °C) temperature conditions, with final cell numbers significantly less (1-2 log10 CFU/ml) than those observed with the derivative peptides alone, or nisin A. To our knowledge, this study is the first investigation that combines bioengineered bacteriocins with the aim of discovering a combination with enhanced antimicrobial activity.


Assuntos
Antibacterianos/metabolismo , Antibacterianos/farmacologia , Lactococcus lactis/metabolismo , Listeria/efeitos dos fármacos , Nisina/metabolismo , Nisina/farmacologia , Antibacterianos/química , Bioengenharia , Lactococcus lactis/genética , Listeria/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana , Nisina/química , Nisina/genética
20.
Cells ; 10(2)2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33670309

RESUMO

Clostridioides difficile is the leading cause of antibiotic-associated diarrhea but can also result in more serious, life-threatening conditions. The incidence of C. difficile infections in hospitals is increasing, both in frequency and severity, and antibiotic-resistant C. difficile strains are advancing. Against this background antimicrobial peptides (AMPs) are an interesting alternative to classic antibiotics. Information on the effects of AMPs on C. difficile will not only enhance the knowledge for possible biomedical application but may also provide insights into mechanisms of C. difficile to adapt or counteract AMPs. This study applies state-of-the-art mass spectrometry methods to quantitatively investigate the proteomic response of C. difficile 630∆erm to sublethal concentrations of the AMP nisin allowing to follow the cellular stress adaptation in a time-resolved manner. The results do not only point at a heavy reorganization of the cellular envelope but also resulted in pronounced changes in central cellular processes such as carbohydrate metabolism. Further, the number of flagella per cell was increased during the adaptation process. The potential involvement of flagella in nisin adaptation was supported by a more resistant phenotype exhibited by a non-motile but hyper-flagellated mutant.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Antibacterianos/farmacologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Nisina/farmacologia , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas de Bactérias/metabolismo , Clostridioides/metabolismo , Clostridioides difficile/metabolismo , Nisina/genética , Nisina/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Proteômica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA