Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 15(727): eadf8366, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-38117900

RESUMO

Graft-versus-host disease (GVHD) remains the major cause of morbidity and nonrelapse mortality (NRM) after hematopoietic cell transplantation (HCT). Inflammatory cytokines mediate damage to key GVHD targets such as intestinal stem cells (ISCs) and also activate receptor interacting protein kinase 1 (RIP1; RIPK1), a critical regulator of apoptosis and necroptosis. We therefore investigated the role of RIP1 in acute GVHD using samples from HCT patients, modeling GVHD damage in vitro with both human and mouse gastrointestinal (GI) organoids, and blocking RIP1 activation in vivo using several well-characterized mouse HCT models. Increased phospho-RIP1 expression in GI biopsies from patients with acute GVHD correlated with tissue damage and predicted NRM. Both the genetic inactivation of RIP1 and the RIP1 inhibitor GNE684 prevented GVHD-induced apoptosis of ISCs in vivo and in vitro. Daily administration of GNE684 for 14 days reduced inflammatory infiltrates in three GVHD target organs (intestine, liver, and spleen) in mice. Unexpectedly, GNE684 administration also reversed the marked loss of regulatory T cells in the intestines and liver during GVHD and reduced splenic T cell exhaustion, thus improving immune reconstitution. Pharmacological and genetic inhibition of RIP1 improved long-term survival without compromising the graft-versus-leukemia (GVL) effect in lymphocytic and myeloid leukemia mouse models. Thus, RIP1inhibition may represent a nonimmunosuppressive treatment for GVHD.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Reconstituição Imune , Leucemia , Humanos , Camundongos , Animais , Citocinas , Leucemia/terapia
2.
Transplant Cell Ther ; 28(8): 472.e1-472.e11, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35643350

RESUMO

Conditioning regimens used for hematopoietic stem cell transplantation (HCT) can escalate the severity of acute T cell-mediated graft-versus-host disease (GVHD) by disrupting gastrointestinal integrity and initiating lipopolysaccharide (LPS)-dependent innate immune cell activation. Activation of the complement cascade has been associated with murine GVHD, and previous work has shown that alternative pathway complement activation can amplify T cell immunity. Whether and how mannan-binding lectin (MBL), a component of the complement system that binds mannose as well as oligosaccharide components of LPS and lipoteichoic acid, affects GVHD is unknown. In this study, we tested the hypothesis that MBL modulates murine GVHD and examined the mechanisms by which it does so. We adoptively transferred C3.SW bone marrow (BM) cells ± T cells into irradiated wild type (WT) or MBL-deficient C57Bl/6 (B6) recipients with or without inhibiting MBL-initiated complement activation using C1-esterase inhibitor (C1-INH). We analyzed the clinical severity of disease expression and analyzed intestinal gene and cell infiltration. In vitro studies assessed MBL expression on antigen-presenting cells (APCs) and compared LPS-induced responses of WT and MBL-deficient APCs. MBL-deficient recipients of donor BM ± T cells exhibited significantly less weight loss over the first 2 weeks post-transplantation weeks compared with B6 controls (P < .05), with similar donor engraftment in the 2 groups. In recipients of C3.SW BM + T cells, the clinical expression of GVHD was less severe (P < .05) and overall survival was better (P < .05) in MBL-deficient mice compared with WT mice. On day-7 post-transplantation, analyses showed that the MBL-deficient recipients exhibited less intestinal IL1b, IL17, and IL12 p40 gene expression (P < .05 for each) and fewer infiltrating intestinal CD11c+, CD11b+, and F4/80+ cells and TCRß+, CD4+, CD4+IL17+, and CD8+ T cells (P < .05 for each). Ovalbumin or allogeneic cell immunizations induced equivalent T cell responses in MBL-deficient and WT mice, demonstrating that MBL-deficiency does not directly impact T cell immunity in the absence of irradiation conditioning. Administration of C1-INH did not alter the clinical expression of GVHD in preconditioned WT B6 recipients, suggesting that MBL amplifies clinical expression of GVHD via a complement-independent mechanism. WT, but not MBL-deficient, APCs express MBL on their surfaces. LPS-stimulated APCs from MBL-deficient mice produced less proinflammatory cytokines (P < .05) and induced weaker alloreactive T cell responses (P < .05) compared with WT APCs. Together, our data show that MBL modulates murine GVHD, likely by amplifying complement-independent, LPS-initiated gastrointestinal inflammation. The results suggest that devising strategies to block LPS/MBL ligation on APCs has the potential to reduce the clinical expression of GVHD.


Assuntos
Doença Enxerto-Hospedeiro , Inflamação , Lectina de Ligação a Manose , Animais , Transplante de Medula Óssea , Linfócitos T CD8-Positivos , Doença Enxerto-Hospedeiro/genética , Inflamação/etiologia , Inflamação/genética , Lipopolissacarídeos/efeitos adversos , Lipopolissacarídeos/farmacologia , Lectina de Ligação a Manose/genética , Camundongos , Camundongos Endogâmicos C57BL , Transplante Homólogo
3.
J Clin Invest ; 132(12)2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35703182

RESUMO

Approximately half of patients with hematologic malignancy who are treated with allogeneic hematopoietic stem cell transplantation (alloHCT) experience graft-versus-host disease (GVHD), which has high mortality rates despite immunosuppressive therapy. IL-12 is known to drive donor T cells toward an inflammatory Th1 lineage in GVHD, but other mechanisms also promote pathological Th1 alloimmune responses. In this issue of the JCI, Dwyer et al. report on their use of transgenic mice and alloHCT models of GVHD to demonstrate that IL-33 acts directly on donor T cells to increase Tbet expression independently of IL-12. Notably, IL-33 amplified T cell receptor-signaling pathways and inhibited production of regulatory molecules. These findings firmly establish IL-33 as an important costimulatory molecule for Th1 cells during GVHD and provide a target for reducing GVHD, especially in the gastrointestinal (GI) tract, where damage drives mortality.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Animais , Doença Enxerto-Hospedeiro/genética , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Interleucina-12 , Interleucina-33/genética , Camundongos , Transplante Homólogo
4.
Front Immunol ; 12: 602067, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33732234

RESUMO

C-type lectin receptors (CLRs) are pattern recognition receptors that are crucial in the innate immune response. The gastrointestinal tract contributes significantly to the maintenance of immune homeostasis; it is the shelter for billions of microorganisms including many genera of Lactobacillus sp. Previously, it was shown that host-CLR interactions with gut microbiota play a crucial role in this context. The Macrophage-inducible C-type lectin (Mincle) is a Syk-coupled CLR that contributes to sensing of mucosa-associated commensals. In this study, we identified Mincle as a receptor for the Surface (S)-layer of the probiotic bacteria Lactobacillus brevis modulating GM-CSF bone marrow-derived cells (BMDCs) functions. We found that the S-layer/Mincle interaction led to a balanced cytokine response in BMDCs by triggering the release of both pro- and anti-inflammatory cytokines. In contrast, BMDCs derived from Mincle-/-, CARD9-/- or conditional Syk-/- mice failed to maintain this balance, thus leading to an increased production of the pro-inflammatory cytokines TNF and IL-6, whereas the levels of the anti-inflammatory cytokines IL-10 and TGF-ß were markedly decreased. Importantly, this was accompanied by an altered CD4+ T cell priming capacity of Mincle-/- BMDCs resulting in an increased CD4+ T cell IFN-γ production upon stimulation with L. brevis S-layer. Our results contribute to the understanding of how commensal bacteria regulate antigen-presenting cell (APC) functions and highlight the importance of the Mincle/Syk/Card9 axis in APCs as a key factor in host-microbiota interactions.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células da Medula Óssea/imunologia , Proteínas Adaptadoras de Sinalização CARD/imunologia , Lectinas Tipo C/imunologia , Levilactobacillus brevis/imunologia , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana/imunologia , Transdução de Sinais/imunologia , Quinase Syk/imunologia , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Linfócitos T CD4-Positivos/imunologia , Citocinas/genética , Citocinas/imunologia , Levilactobacillus brevis/genética , Lectinas Tipo C/genética , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Quinase Syk/genética
5.
Biochem Soc Trans ; 47(6): 1569-1579, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31724699

RESUMO

Bacterial surfaces are rich in glycoconjugates that are mainly present in their outer layers and are of great importance for their interaction with the host innate immune system. The innate immune system is the first barrier against infection and recognizes pathogens via conserved pattern recognition receptors (PRRs). Lectins expressed by innate immune cells represent an important class of PRRs characterized by their ability to recognize carbohydrates. Among lectins in innate immunity, there are three major classes including the galectins, siglecs, and C-type lectin receptors. These lectins may contribute to initial recognition of bacterial glycans, thus providing an early defence mechanism against bacterial infections, but they may also be exploited by bacteria to escape immune responses. In this review, we will first exemplify bacterial glycosylation systems; we will then describe modes of recognition of bacterial glycans by lectins in innate immunity and, finally, we will briefly highlight how bacteria have found ways to exploit these interactions to evade immune recognition.


Assuntos
Imunidade Inata , Lectinas/metabolismo , Polissacarídeos/metabolismo , Animais , Bactérias/metabolismo , Glicosilação , Humanos , Ligação Proteica
6.
Front Microbiol ; 10: 810, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31040840

RESUMO

Alphaviruses and flaviviruses are important human pathogens that include Chikungunya virus (CHIKV), Dengue virus (DENV), and Zika virus (ZIKV), which can cause diseases in humans ranging from arthralgia to hemorrhagic fevers and microcephaly. It was previously shown that treatment with surface layer (S-layer) protein, present on the bacterial cell-envelope of Lactobacillus acidophilus, is able to inhibit viral and bacterial infections by blocking the pathogen's interaction with DC-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN), a trans-membrane protein that is a C-type calcium-dependent lectin. DC-SIGN is known to act as an attachment factor for several viruses including alphaviruses and flaviviruses. In the present study, we used alphaviruses as a model system to dissect the mechanism of S-layer inhibition. We first evaluated the protective effect of S-layer using 3T3 cells, either wild type or stably expressing DC-SIGN, and infecting with the alphaviruses Semliki Forest virus (SFV) and CHIKV and the flaviviruses ZIKV and DENV. DC-SIGN expression significantly enhanced infection by all four viruses. Treatment of the cells with S-layer prior to infection decreased infectivity of all viruses only in cells expressing DC-SIGN. In vitro ELISA experiments showed a direct interaction between S-layer and DC-SIGN; however, confocal microscopy and flow cytometry demonstrated that S-layer binding to the cells was independent of DC-SIGN expression. S-layer protein prevented SFV binding and internalization in DC-SIGN-expressing cells but had no effect on virus binding to DC-SIGN-negative cells. Inhibition of virus binding occurred in a time-dependent manner, with a significant reduction of infection requiring at least a 30-min pre-incubation of S-layer with DC-SIGN-expressing cells. These results suggest that S-layer has a different mechanism of action compared to mannan, a common DC-SIGN-binding compound that has an immediate effect in blocking viral infection. This difference could reflect slower kinetics of S-layer binding to the DC-SIGN present at the plasma membrane (PM). Alternatively, the S-layer/DC-SIGN interaction may trigger the activation of signaling pathways that are required for the inhibition of viral infection. Together our results add important information relevant to the potential use of L. acidophilus S-layer protein as an antiviral therapy.

7.
Genome Announc ; 6(7)2018 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-29449405

RESUMO

Lactobacillus helveticus is a lactic acid bacterium used traditionally in the dairy industry, especially in the manufacture of cheeses. We present here the 2,141,841-bp draft genome sequence of L. helveticus strain ATCC 12046, a potential starter strain for improving cheese production.

8.
Sci Rep ; 7: 41495, 2017 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-28128333

RESUMO

Helicobacter pylori chronically colonises half of the world's human population and is the main cause of ulcers and gastric cancers. Its prevalence and the increase in antibiotic resistance observed recently reflect the high genetic adaptability of this pathogen. Together with high mutation rates and an efficient DNA recombination system, horizontal gene transfer through natural competence makes of H. pylori one of the most genetically diverse bacteria. We show here that transformation capacity is enhanced in strains defective for recN, extending previous work with other homologous recombination genes. However, inactivation of either mutY or polA has no effect on DNA transformation, suggesting that natural competence can be boosted in H. pylori by the persistence of DNA breaks but not by enhanced mutagenesis. The transformation efficiency of the different DNA repair impaired strains correlates with the number of transforming DNA foci formed on the cell surface and with the expression of comB8 and comB10 competence genes. Overexpression of the comB6-B10 operon is sufficient to increase the transformation capacity of a wild type strain, indicating that the ComB complex, present in the bacterial wall and essential for DNA uptake, can be a limiting factor for transformation efficiency.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Helicobacter pylori/genética , Proteínas de Bactérias/genética , Dano ao DNA , Reparo do DNA , DNA Bacteriano/genética , Helicobacter pylori/metabolismo , Mutação/genética , Óperon/genética , Transformação Genética
9.
Int J Biol Macromol ; 92: 998-1005, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27498415

RESUMO

Many species of Lactobacillus sp. possess Surface(s) layer proteins in their envelope. Among other important characteristics S-layer from Lactobacillus acidophilus binds to the cellular receptor DC-SIGN (Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin; CD209), which is involved in adhesion and infection of several families of bacteria. In this report we investigate the activity of new S-layer proteins from the Lactobacillus family (Lactobacillus acidophilus, Lactobacillus brevis, Lactobacillus helveticus and Lactobacillus kefiri) over the infection of representative microorganisms important to human health. After the treatment of DC-SIGN expressing cells with these proteins, we were able to diminish bacterial infection by up to 79% in both gram negative and mycobacterial models. We discovered that pre-treatment of the bacteria with S-layers from Lactobacillus acidophilus and Lactobacillus brevis reduced bacteria viability but also prevent infection by the pathogenic bacteria. We also proved the importance of the glycosylation of the S-layer from Lactobacillus kefiri in the binding to the receptor and thus inhibition of infection. This novel characteristic of the S-layers proteins may contribute to the already reported pathogen exclusion activity for these Lactobacillus probiotic strains; and might be also considered as a novel enzymatic antimicrobial agents to inhibit bacterial infection and entry to host cells.


Assuntos
Infecções Bacterianas/tratamento farmacológico , Moléculas de Adesão Celular/metabolismo , Lactobacillus/química , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/uso terapêutico , Receptores de Superfície Celular/metabolismo , Células 3T3 , Animais , Antibacterianos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Glicosilação/efeitos dos fármacos , Humanos , Glicoproteínas de Membrana/farmacologia , Camundongos
10.
Appl Microbiol Biotechnol ; 100(19): 8475-84, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27376794

RESUMO

In this work, we studied the role of surface layer (S-layer) proteins in the adaptation of Lactobacillus acidophilus ATCC 4356 to the osmotic stress generated by high salt. The amounts of the predominant and the auxiliary S-layer proteins SlpA and SlpX were strongly influenced by the growth phase and high-salt conditions (0.6 M NaCl). Changes in gene expression were also observed as the mRNAs of the slpA and slpX genes increased related to the growth phase and presence of high salt. A growth stage-dependent modification on the S-layer protein profile in response to NaCl was observed: while in control conditions, the auxiliary SlpX protein represented less than 10 % of the total S-layer protein, in high-salt conditions, it increased to almost 40 % in the stationary phase. The increase in S-layer protein synthesis in the stress condition could be a consequence of or a way to counteract the fragility of the cell wall, since a decrease in the cell wall thickness and envelope components (peptidoglycan layer and lipoteichoic acid content) was observed in L. acidophilus when compared to a non-S-layer-producing species such as Lactobacillus casei. Also, the stationary phase and growth in high-salt medium resulted in increased release of S-layer proteins to the supernatant medium. Overall, these findings suggest that pre-growth in high-salt conditions would result in an advantage for the probiotic nature of L. acidophilus ATCC 4356 as the increased amount and release of the S-layer might be appropriate for its antimicrobial capacity.


Assuntos
Expressão Gênica , Lactobacillus acidophilus/genética , Lactobacillus acidophilus/metabolismo , Glicoproteínas de Membrana/metabolismo , Pressão Osmótica , Lactobacillus acidophilus/efeitos dos fármacos , Cloreto de Sódio/metabolismo
11.
Genome Announc ; 3(1)2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25593259

RESUMO

We present the 1,956,699-bp draft genome sequence of Lactobacillus acidophilus strain ATCC 4356. Comparative genomic analysis revealed 99.96% similarity with L. acidophilus NCFM NC_006814.3 and 99.97% with La-14 NC_021181.2 genomes.

12.
PLoS One ; 9(10): e111114, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25354162

RESUMO

Lysinibacillus sphaericus strains belonging the antigenic group H5a5b produce spores with larvicidal activity against larvae of Culex mosquitoes. C7, a new isolated strain, which presents similar biochemical characteristics and Bin toxins in their spores as the reference strain 2362, was, however, more active against larvae of Culex mosquitoes. The contribution of the surface layer protein (S-layer) to this behaviour was envisaged since this envelope protein has been implicated in the pathogenicity of several bacilli, and we had previously reported its association to spores. Microscopic observation by immunofluorescence detection with anti S-layer antibody in the spores confirms their attachment. S-layers and BinA and BinB toxins formed high molecular weight multimers in spores as shown by SDS-PAGE and western blot detection. Purified S-layer from both L. sphaericus C7 and 2362 strain cultures was by itself toxic against Culex sp larvae, however, that from C7 strain was also toxic against Aedes aegypti. Synergistic effect between purified S-layer and spore-crystal preparations was observed against Culex sp. and Aedes aegypti larvae. This effect was more evident with the C7 strain. In silico analyses of the S-layer sequence suggest the presence of chitin-binding and hemolytic domains. Both biochemical characteristics were detected for both S-layers strains that must justify their contribution to pathogenicity.


Assuntos
Aedes/efeitos dos fármacos , Bacillaceae/química , Culex/efeitos dos fármacos , Glicoproteínas de Membrana/toxicidade , Sequência de Aminoácidos , Animais , Quitina/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/farmacologia , Dados de Sequência Molecular , Ligação Proteica , Multimerização Proteica , Esporos Bacterianos/química
13.
Biochem Biophys Res Commun ; 422(4): 590-5, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22595457

RESUMO

It has been previously described that S-layer binds to the C-type lectin DC-specific ICAM-3-grabbing nonintegrin (DC-SIGN, CD209). It was also shown that DC-SIGN is a cell-surface adhesion factor that enhances viral entry of several virus families. Among those, Junin virus (JUNV) entry is enhanced in cells expressing DC-SIGN and for that reason surface-layer protein (S-layer) of Lactobacillus acidophilus ATCC 4365 was evaluated as a possible JUNV inhibitor. Experiments using 3T3 cells stably expressing DC-SIGN, showed an almost complete inhibition of JUNV infection when they were treated with S-layer in a similar extend as the inhibition shown by mannan. However no inhibition effect was observed in 3T3 wild type cells or in 3T3 cells expressing liver/lymph node-specific ICAM-3 grabbing nonintegrin (L-SIGN or DC-SIGNR or CD209L). Treatments with S-layer during different times in the infection demonstrated that inhibition was only observed when S-layer was presented in early stages of the viral infection. This inhibition does not involve the classic recognition of mannose by this C-type lectin as the S-layer showed no evidence to be glycosylated. In fact, the highly basic nature of the S-layer (pI>9.5) seems to be involved in electrostatic interactions between DC-SIGN and S-layer, since high pH abolished the inhibitory effect on infection cause by the S-layer. In silico analysis predicts a Ca(2+)-dependant carbohydrate recognition domain in the SlpA protein. This novel characteristic of the S-layer, a GRAS status protein, contribute to the pathogen exclusion reported for this probiotic strain and may be applied as an antiviral agent to inhibit several kinds of viruses.


Assuntos
Antivirais/farmacologia , Proteínas de Bactérias/farmacologia , Vírus Junin/efeitos dos fármacos , Lactobacillus acidophilus , Glicoproteínas de Membrana/farmacologia , Internalização do Vírus/efeitos dos fármacos , Células 3T3 , Sequência de Aminoácidos , Animais , Antivirais/química , Antivirais/isolamento & purificação , Infecções por Arenaviridae , Proteínas de Bactérias/isolamento & purificação , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Chlorocebus aethiops , Sequência Consenso , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/isolamento & purificação , Camundongos , Dados de Sequência Molecular , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Células Vero
14.
J Microbiol Biotechnol ; 21(2): 147-53, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21364296

RESUMO

Bacillus species have been involved in metal association as biosorbents, but there is not a clear understanding of this chelating property. In order to evaluate this metal chelating capacity, cultures and spores from Grampositive bacteria of species either able or unable to produce surface layer proteins (S-layers) were analyzed for their capacity of copper biosorption. Only those endowed of S-layers, like Bacillus sphaericus and B. thuringiensis, showed a significant biosorption capacity. This capacity (nearly 50%) was retained after heating of cultures, thus supporting that structural elements of the envelopes are responsible for such activity. Purified Slayers from two Bacillus sphaericus strains had the ability to biosorb copper. Copper biosorption parameters were determined for strain B. sphaericus 2362, and after analyses by means of the Langmuir model, the affinity and capacity were shown to be comparable to other bacterial biosorbents. A competitive effect of Ca2+ and Zn2+, but not of Cd2+, was also observed, thus indicating that other cations may be biosorbed by this protein. Spores that have been shown to be proficient for copper biosorption were further analyzed for the presence of Slayer content. The retention of S-layers by these spores was clearly observed, and after extensive treatment to eliminate the S-layers, the biosorption capacity of these spores was significantly reduced. For the first time, a direct correlation between S-layer protein content and metal biosorption capacity is shown. This capacity is linked to the retention of S-layer proteins attached to Bacillus spores and cells.


Assuntos
Bacillus/metabolismo , Glicoproteínas de Membrana/metabolismo , Metais/metabolismo , Cátions Bivalentes/metabolismo , Quelantes/metabolismo , Ligação Proteica , Esporos Bacterianos/metabolismo
15.
J Microbiol Methods ; 83(2): 164-7, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20807556

RESUMO

We here describe a new method for electroporation of Lactobacillus species, obligately homofermentative and facultatively heterofermentative, based on the cell-wall weakening resulting from growth in high-salt media. For L. casei, optimum transformation efficiency of up to 10(5) transformants per microgram of plasmid DNA was achieved following growth in the presence of 0.9 M NaCl. Plasmids of different sizes and replication origins were also similarly transformed. These competent cells could be used either directly or stored frozen, up to 1 month, for future use, with similar efficiency. This protocol was assayed with different Lactobacillus species: L. delbrueckii subsp. lactis, L. paracasei, L. plantarum and L. acidophilus, and it was found that they were transformed with similar efficiency.


Assuntos
Meios de Cultura/química , Eletroporação/métodos , Lactobacillus/genética , Sais/metabolismo , Criopreservação/métodos , Lactobacillus/crescimento & desenvolvimento , Viabilidade Microbiana , Plasmídeos
16.
Appl Environ Microbiol ; 76(3): 974-7, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19948852

RESUMO

We have previously described a murein hydrolase activity for the surface layer (S-layer) of Lactobacillus acidophilus ATCC 4356. Here we show that, in combination with nisin, this S-layer acts synergistically to inhibit the growth of pathogenic Gram-negative Salmonella enterica and potential pathogenic Gram-positive bacteria, Staphylococcus aureus and Bacillus cereus. In addition, bacteriolytic effects were observed for the Gram-positive species tested. We postulate that the S-layer enhances the access of nisin into the cell membrane by enabling it to cross the cell wall, while nisin provides the sudden ion-nonspecific dissipation of the proton motive force required to enhance the S-layer murein hydrolase activity.


Assuntos
Antibacterianos/farmacologia , Conservantes de Alimentos/farmacologia , Lactobacillus acidophilus/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/farmacologia , Nisina/farmacologia , Bacillus cereus/efeitos dos fármacos , Bacillus cereus/crescimento & desenvolvimento , Membrana Celular/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Contagem de Colônia Microbiana , Sinergismo Farmacológico , Microbiologia de Alimentos , Genes Bacterianos/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Bactérias Gram-Positivas/crescimento & desenvolvimento , Temperatura Alta , Testes de Sensibilidade Microbiana , Permeabilidade , Polilisina/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Tensoativos/farmacologia
17.
Appl Environ Microbiol ; 74(24): 7824-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18931300

RESUMO

We describe a new enzymatic functionality for the surface layer (S-layer) of Lactobacillus acidophilus ATCC 4356, namely, an endopeptidase activity against the cell wall of Salmonella enterica serovar Newport, assayed via zymograms and identified by Western blotting. Based on amino acid sequence comparisons, the hydrolase activity was predicted to be located at the C terminus. Subsequent cloning and expression of the C-terminal domain in Bacillus subtilis resulted in the functional verification of the enzymatic activity.


Assuntos
Lactobacillus acidophilus/enzimologia , Glicoproteínas de Membrana/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Sequência de Aminoácidos , Bacillus subtilis/enzimologia , Parede Celular/metabolismo , Clonagem Molecular , Dados de Sequência Molecular , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...