Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Digestion ; 103(4): 269-286, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35184054

RESUMO

INTRODUCTION: Gut microbiota alterations cause inflammation in patients with ulcerative colitis (UC). Fecal microbiota transplantation (FMT) enables manipulating the microbiota's composition, but the mechanisms underlying colonization of the posttransplantation microbiota are poorly understood. METHODS: In this open-label, nonrandomized study, the FMT efficacy and changes in the gut microbiota were evaluated in 8 UC patients with mild-to-moderately active endoscopic colonic lesions. Compositional changes in the fecal and mucosal microbiotas between donors and recipients were examined via 16S rRNA-based sequencing. To investigate the effects of oral corticosteroids on microbiota colonization, FMT was performed in germ-free prednisolone (PSL)-administered mice to examine the factors determining colonization. RESULTS: Four UC patients achieved clinical remission (CR) after FMT, and 3 also achieved endoscopic remission. The fecal microbiotas of the CR patients changed similar to those of the donors after FMT. The mucin-coding gene, MUC2, was less expressed in the colons of the PSL-dependent patients than in the PSL-free patients. In the mice, PSL treatment decreased the fecal mucin production and altered the posttransplantation fecal microbiota composition. Adding either exogenous mucin or the mucin secretagogue, rebamipide, partially alleviated the PSL-induced dysbiosis of the gut microbiota. Administering rebamipide with FMT from healthy donors relieved inflammation in mice with Enterococcus faecium-induced colitis. CONCLUSION: Colonic mucin controlled the gut microbiota composition, and oral corticosteroid treatment modified the gut microbiota partly by reducing the colonic mucin.


Assuntos
Colite Ulcerativa , Microbiota , Corticosteroides , Animais , Colite Ulcerativa/terapia , Fezes , Inflamação , Camundongos , Mucinas , RNA Ribossômico 16S/genética , Resultado do Tratamento
2.
J Infect Chemother ; 28(5): 651-656, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35078721

RESUMO

INTRODUCTION: Clostridioides difficile (C. difficile) produces three kinds of toxins: toxin A (enterotoxin), toxin B (cytotoxin), and C. difficile transferase (CDT), a binary toxin. Some strains show positivity only for toxin B. These strains reportedly possess a gene for toxin A, tcdA. However, toxin A production is inhibited due to a mutated stop codon and/or deletion within the tcdA gene. Here for the first case in Japan, we describe toxin genomes and proteins of a strain possessing only toxin B and lacking a complete tcdA gene, along with clinical manifestations. METHODS: C. difficile was isolated from the bloody stool of a 60-year-old female patient treated with meropenem. Although a rapid detection kit of toxins (C. DIFF QUIK CHEK COMPLETE®, TechLab, Blacksburg, VA, USA) showed positivity, Western blotting detected no toxins. Therefore, we explored the strain's toxin genes and their sequences to determine whether the strain possessed a toxin. RESULTS: Polymerase chain reaction did not identify toxin genes. Whole-genome sequencing analysis showed that a gene for toxin A, tcdA, was completely deleted in the strain. Moreover, 701 mutations and some deletions/insertions were identified on the tcdB gene. CONCLUSIONS: We isolated a rare strain of C. difficile producing only toxin B and lacking a complete tcdA gene herein Japan. The possibility of a false negative needs to be considered with a genetic method for a diagnose of C. difficile infection.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Proteínas de Bactérias/análise , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Clostridioides , Clostridioides difficile/genética , Enterotoxinas/genética , Feminino , Humanos , Japão , Pessoa de Meia-Idade
3.
Microbiol Immunol ; 65(10): 432-437, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34219270

RESUMO

Clostridium botulinum causes infant and adult intestinal botulism by colonizing in the intestine and producing botulinum neurotoxin (BoNT). Antimicrobial agents are not currently used for treatment due to the potential facilitation of BoNT production and bacterial cell lysis, which releases toxins into the intestinal lumen. In this study, we analyzed effects of four antibiotics on the viability of and BoNT production by four C. botulinum group I strains. Our results indicate that metronidazole rapidly reduced their viability without enhancing BoNT production. Antibiotics with these properties may promote elimination of C. botulinum from the intestines while maintaining low levels of BoNT.


Assuntos
Toxinas Botulínicas , Botulismo , Clostridium botulinum , Antibacterianos/farmacologia , Humanos
4.
Microbiol Immunol ; 62(2): 80-89, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29266585

RESUMO

Hemagglutinin (HA) is one of the components of botulinum neurotoxin (BoNT) complexes and it promotes the absorption of BoNT through the intestinal epithelium by at least two specific mechanisms: cell surface attachment by carbohydrate binding, and epithelial barrier disruption by E-cadherin binding. It is known that HA forms a three-arm structure, in which each of three protomers has three carbohydrate-binding sites and one E-cadherin-binding site. A three-arm form of HA is considered to bind to these ligands simultaneously. In the present study, we investigated how the multivalency effect of HA influences its barrier-disrupting activity. We prepared type B full-length HA (three-arm form) and mini-HA, which is a deletion mutant lacking the trimer-forming domain. Size-exclusion chromatography analysis showed that mini-HA exists as dimers (two-arm form) and monomers (one-arm form), which are then separated. We examined the multivalency effect of HA on the barrier-disrupting activity, the E-cadherin-binding activity, and the attachment activity to the basolateral cell surface. Our results showed that HA initially attaches to the basal surface of Caco-2 cells by carbohydrate binding and then moves to the lateral cell surface, where the HA acts to disrupt the epithelial barrier. Our results showed that the multivalency effect of HA enhances the barrier-disrupting activity in Caco-2 cells. We found that basal cell surface attachment and binding ability to immobilized E-cadherin were enhanced by the multivalency effect of HA. These results suggest that at least these two factors induced by the multivalency effect of HA cause the enhancement of the barrier-disrupting activity.


Assuntos
Toxinas Botulínicas Tipo A/metabolismo , Células Epiteliais/metabolismo , Hemaglutininas/metabolismo , Mucosa Intestinal/metabolismo , Antígenos CD , Sítios de Ligação , Toxinas Botulínicas/química , Toxinas Botulínicas Tipo A/química , Células CACO-2 , Caderinas/química , Caderinas/metabolismo , Carboidratos , Clostridium botulinum tipo B/genética , DNA Bacteriano/genética , Hemaglutininas/química , Hemaglutininas/genética , Humanos , Absorção Intestinal , Mutagênese Sítio-Dirigida , Plasmídeos , Ligação Proteica , Proteínas Recombinantes , Deleção de Sequência
5.
Jpn J Infect Dis ; 77(1): 16-20, 2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-37648491

RESUMO

Equine botulinum antitoxin is one of the most popular countermeasures for human botulism. The unitage of the antitoxin product is defined according to national minimum requirement or pharmacopoeia in each country by referring to national standard antitoxins for four types (A, B, E, and F). With the expected depletion of the national standard antitoxins, replacement national standard antitoxins are produced and standardized through collaboration of the National Control Laboratory and other participants, including manufacturer(s). Therefore, Japanese National Standard Botulinum Antitoxin Type A, Equine, was replaced according to the results of a collaborative study involving the National Institute of Infectious Diseases and KM Biologics Co., Ltd. The unitage of the replacement material was determined through mouse neutralization tests, which involved toxin-antitoxin mixture injection at pH 7.0. Potency value of 440 units/vial was obtained. However, the Japanese Minimum Requirement for Biological Products was revised, and the neutralization reactions were repeated at pH 6.0, for which considerably different potency value (656 units/vial) and survival profile of mice were obtained. In September 2021, the replacement material, Japanese National Standard Botulinum Antitoxin Type A, Equine, lot 2, was established with potency value of 656 Units/vial. The impact of pH-dependent change in potency on antitoxin quality control is discussed.


Assuntos
Antitoxinas , Toxinas Botulínicas Tipo A , Botulismo , Animais , Cavalos , Humanos , Camundongos , Antitoxina Botulínica/uso terapêutico , Japão , Botulismo/tratamento farmacológico , Botulismo/veterinária , Padrões de Referência
6.
J Gastroenterol ; 57(10): 770-783, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35882645

RESUMO

BACKGROUND: Dysbiosis of gut microbiota promotes colitis in ulcerative colitis (UC). Enterococcus faecium is an important constituent of dysbiotic microbiota. However, the mechanisms underlying E. faecium-induced colitis remain unclear. METHODS: Overall, 23 E. faecium strains isolated from human feces and 3 commercial strains were inoculated into Il10-/- mice. Mouse colons were histologically evaluated and analyzed using real-time PCR analysis of cytokines. Genes in 26 E. faecium strains were identified by whole-genome shotgun sequencing of genomic DNA. The production of reactive oxygen species (ROS) from each strain was measured. An antioxidant, lipoic acid, was orally administered to the colitis mouse model. RESULTS: Inoculation of E. faecium derived from patients with UC resulted in colitis in Il10-/- mice. The genotypes of 26 strains were characterized by identifying 1893 known genes; clustering all the strains based on the genotypes showed two major groups-inflammatory and probiotic clusters. Additionally, linear discriminant analysis clarified that lipoic acid metabolism was a significantly abundant pathway in the probiotic cluster compared to the inflammatory cluster. Further, the production of ROS was greater in inflammatory than in probiotic strains. Administration of lipoic acid in E. faecium-inoculated mice ameliorated colitis. CONCLUSIONS: Enterococcus faecium strains in the inflammatory cluster promoted colitis with higher production of ROS than the strains in the probiotic cluster.


Assuntos
Colite Ulcerativa , Colite , Enterococcus faecium , Probióticos , Animais , Antioxidantes , Colite/induzido quimicamente , Disbiose , Enterococcus faecium/patogenicidade , Genótipo , Humanos , Interleucina-10/genética , Camundongos , Espécies Reativas de Oxigênio , Ácido Tióctico/farmacologia , Ácido Tióctico/uso terapêutico
7.
Front Microbiol ; 13: 720308, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35185840

RESUMO

Clostridium botulinum produces botulinum neurotoxin complexes that cause botulism. Previous studies elucidated the molecular pathogenesis of botulinum neurotoxin complexes; however, it currently remains unclear whether other components of the bacterium affect host cells. Recent studies provided insights into the role of bacterial membrane vesicles (MVs) produced by some bacterial species in host immunity and pathology. We herein examined and compared the cellular effects of MVs isolated from four strains of C. botulinum with those of closely related Clostridium sporogenes and two strains of the symbiont Clostridium scindens. MVs derived from all strains induced inflammatory cytokine expression in intestinal epithelial and macrophage cell lines. Cytokine expression was dependent on myeloid differentiation primary response (MyD) 88 and TIR-domain-containing adapter-inducing interferon-ß (TRIF), essential adaptors for toll-like receptors (TLRs), and TLR1/2/4. The inhibition of actin polymerization impeded the uptake of MVs in RAW264.7 cells, however, did not reduce the induction of cytokine expression. On the other hand, the inhibition of dynamin or phosphatidylinositol-3 kinase (PI3K) suppressed the induction of cytokine expression by MVs, suggesting the importance of these factors downstream of TLR signaling. MVs also induced expression of Reg3 family antimicrobial peptides via MyD88/TRIF signaling in primary cultured mouse small intestinal epithelial cells (IECs). The present results indicate that MVs from C. botulinum and related clostridial species induce host innate immune responses.

8.
Phytother Res ; 25(11): 1707-13, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21721062

RESUMO

trans-Anethole (anethole), a major component of anise oil, has a broad antimicrobial spectrum with antimicrobial activity relatively weaker than those of well-known antibiotics, and significantly enhances the antifungal activity of polygodial and dodecanol against the baker's yeast Saccharomyces cerevisiae and human pathogenic yeast Candida albicans. However, the antifungal mechanism of anethole is unresolved. Anethole demonstrated antifungal activity against the filamentous fungus, Mucor mucedo IFO 7684, accompanied by hyphal morphological changes such as swollen hyphae at the tips. Its minimum growth inhibitory concentration was 0.625 mM. A hyperosmotic condition (1.2 M sorbitol) restricted the induction of morphological changes, while hypoosmotic treatment (distilled water) induced bursting of hyphal tips and leakage of cytoplasmic constituents. Furthermore, anethole dose-dependently inhibited chitin synthase (CHS) activity in permeabilized hyphae in an uncompetitive manner. These results suggest that the morphological changes of M. mucedo could be explained by the fragility of cell walls caused by CHS inhibition.


Assuntos
Anisóis/farmacologia , Antifúngicos/farmacologia , Quitina Sintase/metabolismo , Proteínas Fúngicas/metabolismo , Mucor/efeitos dos fármacos , Derivados de Alilbenzenos , Permeabilidade da Membrana Celular , Parede Celular/efeitos dos fármacos , Hifas/efeitos dos fármacos , Hifas/enzimologia , Testes de Sensibilidade Microbiana , Mucor/enzimologia
9.
Gen Physiol Biophys ; 30(1): 106-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21460419

RESUMO

Sporulation of the yeast Saccharomyces cerevisiae is negatively regulated by cyclic AMP (cAMP). This microbial cell differentiation process was applied for the screening of a substance that can elevate the intracellular cAMP level. Among nucleoside 5'-alkylphosphates, uridine 5'-eicosylphosphate (UMPC20) selectively and predominantly inhibited ascospore formation of the yeast cells. We suppose the inhibitory effect of UMPC20 could indeed reflect the elevation of the cellular cAMP level.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Divisão Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Timidina Monofosfato/análogos & derivados , Uridina Monofosfato/análogos & derivados , Monofosfato de Adenosina/farmacologia , Saccharomyces cerevisiae/metabolismo , Uridina Monofosfato/farmacologia
10.
Nat Cancer ; 2(10): 1039-1054, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-35121877

RESUMO

Gut dysbiosis is observed in chronic hepatobiliary diseases and is frequently associated with liver carcinogenesis; however, the extent and specific mechanisms triggered by alterations in the microbiota mediating tumorigenesis in these patients remain unclear. Here we show that Enterococcus faecalis is abundant in the microbiota of patients with hepatitis C virus-related chronic liver disease. Xenotransplantation of gut microbiota from these patients increased the number of spontaneous liver tumors in mice and enhanced susceptibility to liver carcinogens. Hepatic colonization by gelE-positive E. faecalis increased liver expression of proliferative genes in a TLR4-Myd88-dependent manner, leading to liver tumorigenesis. Moreover, decreased fecal deoxycholic acid levels were associated with colonization by E. faecalis. Overall, these data identify E. faecalis as a key promoter of liver carcinogenesis.


Assuntos
Enterococcus faecalis , Hepatopatias , Animais , Carcinogênese , Disbiose , Enterococcus faecalis/metabolismo , Humanos , Camundongos
11.
Toxins (Basel) ; 12(5)2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32392791

RESUMO

Botulinum neurotoxin (BoNT) is the most potent natural toxin known. Of the seven BoNT serotypes (A to G), types A, B, E, and F cause human botulism. Treatment of human botulism requires the development of effective toxin-neutralizing antibodies without side effects such as serum sickness and anaphylaxis. In this study, we generated fully human monoclonal antibodies (HuMAbs) against serotype B BoNT (BoNT/B1) using a murine-human chimera fusion partner cell line named SPYMEG. Of these HuMAbs, M2, which specifically binds to the light chain of BoNT/B1, showed neutralization activity in a mouse bioassay (approximately 10 i.p. LD50/100 µg of antibody), and M4, which binds to the C-terminal of heavy chain, showed partial protection. The combination of two HuMAbs, M2 (1.25 µg) and M4 (1.25 µg), was able to completely neutralize BoNT/B1 (80 i.p. LD50) with a potency greater than 80 i.p. LD50/2.5 µg of antibodies, and was effective both prophylactically and therapeutically in the mouse model of botulism. Moreover, this combination showed broad neutralization activity against three type B subtypes, namely BoNT/B1, BoNT/B2, and BoNT/B6. These data demonstrate that the combination of M2 and M4 is promising in terms of a foundation for new human therapeutics for BoNT/B intoxication.


Assuntos
Anticorpos Monoclonais/farmacologia , Toxinas Botulínicas Tipo A/antagonistas & inibidores , Botulismo/prevenção & controle , Anticorpos Amplamente Neutralizantes/farmacologia , Clostridium botulinum/efeitos dos fármacos , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Sítios de Ligação de Anticorpos , Toxinas Botulínicas Tipo A/imunologia , Botulismo/imunologia , Botulismo/microbiologia , Anticorpos Amplamente Neutralizantes/imunologia , Clostridium botulinum/imunologia , Modelos Animais de Doenças , Quimioterapia Combinada , Epitopos , Feminino , Humanos , Hibridomas , Camundongos , Testes de Neutralização , Ligação Proteica
12.
Int J Infect Dis ; 91: 22-31, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31740408

RESUMO

OBJECTIVES: Methicillin-resistant Staphylococcus aureus (MRSA) causes hospital- and community-acquired infections. It is not clear whether genetic characteristics of the bacteria contribute to disease pathogenesis in MRSA infection. We hypothesized that whole genome analysis of MRSA strains could reveal the key gene loci and/or the gene mutations that affect clinical manifestations of MRSA infection. METHODS: Whole genome sequences (WGS) of MRSA of 154 strains were analyzed with respect to clinical manifestations and data. Further, we evaluated the association between clinical manifestations in MRSA infection and genomic information. RESULTS: WGS revealed gene mutations that correlated with clinical manifestations of MRSA infection. Moreover, 12 mutations were selected as important mutations by Random Forest analysis. Cluster analysis revealed strains associated with a high frequency of bloodstream infection (BSI). Twenty seven out of 34 strains in this cluster caused BSI. These strains were all positive for collagen adhesion gene (cna) and have mutations in the locus, those were selected by Random Forest analysis. Univariate and multivariate analysis revealed that these gene mutations were the predictor for the incidence of BSI. Interestingly, mutant CNA protein showed lower attachment ability to collagen, suggesting that the mutant protein might contribute to the dissemination of bacteria. CONCLUSIONS: These findings suggest that the bacterial genotype affects the clinical characteristics of MRSA infection.


Assuntos
Adesinas Bacterianas/genética , Bacteriemia/microbiologia , Staphylococcus aureus Resistente à Meticilina/genética , Infecções Estafilocócicas/microbiologia , Adulto , Idoso , DNA Bacteriano , Feminino , Genoma Bacteriano , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Sequenciamento Completo do Genoma
13.
Genome Biol ; 20(1): 252, 2019 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-31767028

RESUMO

BACKGROUND: Recent metagenomic analyses have revealed dysbiosis of the gut microbiota of ulcerative colitis (UC) patients. However, the impacts of this dysbiosis are not fully understood, particularly at the strain level. RESULTS: We perform whole-genome shotgun sequencing of fecal DNA extracts from 13 healthy donors and 16 UC and 8 Crohn's disease (CD) patients. The microbiota of UC and CD patients is taxonomically and functionally divergent from that of healthy donors, with E. faecium being the most differentially abundant species between the two microbial communities. Transplantation of feces from UC or CD patients into Il10-/- mice promotes pathological inflammation and cytokine expression in the mouse colon, although distinct cytokine expression profiles are observed between UC and CD. Unlike isolates derived from healthy donors, E. faecium isolates from the feces of UC patients, along with E. faecium strain ATCC 19434, promotes colitis and colonic cytokine expression. Inflammatory E. faecium strains, including ATCC 19434 and a UC-derived strain, cluster separately from commercially available probiotic strains based on whole-genome shotgun sequencing analysis. The presence of E. faecium in fecal samples is associated with large disease extent and the need for multiple medications in UC patients. CONCLUSIONS: E. faecium strains derived from UC patients display an inflammatory genotype that causes colitis.


Assuntos
Colite Ulcerativa/microbiologia , Enterococcus faecalis/patogenicidade , Microbioma Gastrointestinal , Metagenoma , Animais , Estudos de Casos e Controles , Colite/etiologia , Colite/patologia , Colite Ulcerativa/tratamento farmacológico , Colo/patologia , Doença de Crohn/microbiologia , Modelos Animais de Doenças , Quimioterapia Combinada , Enterococcus faecalis/genética , Transplante de Microbiota Fecal , Fezes/microbiologia , Feminino , Humanos , Interleucina-10/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
14.
FEBS J ; 282(17): 3334-47, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26077172

RESUMO

Botulinum neurotoxin is conventionally divided into seven serotypes, designated A-G, and is produced as large protein complexes through associations with non-toxic components, such as hemagglutinin (HA) and non-toxic non-HA. These non-toxic proteins dramatically enhance the oral toxicity of the toxin complex. HA is considered to have a role in toxin transport through the intestinal epithelium by carbohydrate binding and epithelial barrier-disrupting activity. Type A and B HAs disrupt E-cadherin-mediated cell adhesion, and, in turn, the intercellular epithelial barrier. Type C HA (HA/C) disrupts the barrier function by affecting cell morphology and viability, the mechanism of which remains unknown. In this study, we identified GM3 as the target molecule of HA/C. We found that sialic acid binding of HA is essential for the activity. It was abolished when cells were pre-treated with an inhibitor of ganglioside synthesis. Consistent with this, HA/C bound to a-series gangliosides in a glycan array. In parallel, we isolated clones resistant to HA/C activity from a susceptible mouse fibroblast strain. These cells lacked expression of ST-I, the enzyme that transfers sialic acid to lactosylceramide to yield GM3. These clones became sensitive to HA/C activity when GM3 was expressed by transfection with the ST-I gene. The sensitivity of fibroblasts to HA/C was reduced by expressing ganglioside synthesis genes whose products utilize GM3 as a substrate and consequently generate other a-series gangliosides, suggesting a GM3-specific mechanism. Our results demonstrate that HA/C affects cells in a GM3-dependent manner.


Assuntos
Proteínas de Bactérias/metabolismo , Clostridium botulinum/química , Gangliosídeo G(M3)/metabolismo , Hemaglutininas/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/farmacologia , Caderinas/genética , Caderinas/metabolismo , Sequência de Carboidratos , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Clostridium botulinum/metabolismo , Cães , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Gangliosídeo G(M3)/química , Expressão Gênica , Hemaglutininas/genética , Hemaglutininas/farmacologia , Células Madin Darby de Rim Canino , Camundongos , Análise em Microsséries , Dados de Sequência Molecular , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Sialiltransferases/deficiência , Sialiltransferases/genética
15.
Nat Commun ; 6: 6255, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25687350

RESUMO

To cause food-borne botulism, botulinum neurotoxin (BoNT) in the gastrointestinal lumen must traverse the intestinal epithelial barrier. However, the mechanism by which BoNT crosses the intestinal epithelial barrier remains unclear. BoNTs are produced along with one or more non-toxic components, with which they form progenitor toxin complexes (PTCs). Here we show that serotype A1 L-PTC, which has high oral toxicity and makes the predominant contribution to causing illness, breaches the intestinal epithelial barrier from microfold (M) cells via an interaction between haemagglutinin (HA), one of the non-toxic components, and glycoprotein 2 (GP2). HA strongly binds to GP2 expressed on M cells, which do not have thick mucus layers. Susceptibility to orally administered L-PTC is dramatically reduced in M-cell-depleted mice and GP2-deficient (Gp2(-/-)) mice. Our finding provides the basis for the development of novel antitoxin therapeutics and delivery systems for oral biologics.


Assuntos
Toxinas Botulínicas Tipo A/química , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Intestinos/citologia , Animais , Carboidratos/química , Clostridium botulinum , Células Dendríticas/citologia , Cães , Endocitose , Feminino , Proteínas Ligadas por GPI/metabolismo , Glutationa Transferase/metabolismo , Hemaglutininas/química , Humanos , Mucosa Intestinal/metabolismo , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Neurônios/metabolismo , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/química , Ligação Proteica , Proteínas Recombinantes de Fusão/química
16.
PLoS One ; 9(10): e111170, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25340348

RESUMO

Botulinum neurotoxin (BoNT) inhibits neurotransmitter release in motor nerve endings, causing botulism, a condition often resulting from ingestion of the toxin or toxin-producing bacteria. BoNTs are always produced as large protein complexes by associating with a non-toxic protein, non-toxic non-hemagglutinin (NTNH), and some toxin complexes contain another non-toxic protein, hemagglutinin (HA), in addition to NTNH. These accessory proteins are known to increase the oral toxicity of the toxin dramatically. NTNH has a protective role against the harsh conditions in the digestive tract, while HA is considered to facilitate intestinal absorption of the toxin by intestinal binding and disruption of the epithelial barrier. Two specific activities of HA, carbohydrate and E-cadherin binding, appear to be involved in these processes; however, the exact roles of these activities in the pathogenesis of botulism remain unclear. The toxin is conventionally divided into seven serotypes, designated A through G. In this study, we identified the amino acid residues critical for carbohydrate and E-cadherin binding in serotype B HA. We constructed mutants defective in each of these two activities and examined the relationship of these activities using an in vitro intestinal cell culture model. Our results show that the carbohydrate and E-cadherin binding activities are functionally and structurally independent. Carbohydrate binding potentiates the epithelial barrier-disrupting activity by enhancing cell surface binding, while E-cadherin binding is essential for the barrier disruption.


Assuntos
Caderinas/química , Carboidratos/química , Clostridium botulinum tipo B/química , Hemaglutininas/química , Antígenos CD , Sítios de Ligação , Toxinas Botulínicas/química , Botulismo/microbiologia , Células CACO-2 , Impedância Elétrica , Humanos , Absorção Intestinal , Intestinos/microbiologia , Mucinas/química , Neurotransmissores/química , Plasmídeos , Ligação Proteica
17.
J Antibiot (Tokyo) ; 64(7): 469-74, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21522157

RESUMO

The alkylguanidium chain attached to the polyol lactone ring of niphimycin (NM) is considered a requisite for the fungicidal activity of NM characterized by vacuole membrane fragmentation and oxidative stress induction. The addition of N-methyl-N″-dodecylguanidine to the medium can enhance the vacuole-targeting fungicidal activity of amphotericin B (AmB), in which the lactone ring has no such alkylguanidium chain, on Saccharomyces cerevisiae cells. In this study, the enhancement effect of N-methyl-N″-dodecylguanidine on the vacuole-targeting fungicidal activity of AmB was examined against Candida albicans in RPMI 1640 medium at 37 °C. N-methyl-N″-dodecylguanidine was lethal to C. albicans cells and additionally enhanced the vacuole disruptive activity of AmB against this pathogenic fungus. N-methyl-N″-dodecylguanidine elevated the generation of cellular reactive oxygen species when added alone in a dose-dependent manner, but its enhancement effect on AmB lethality did not accompany amplification of oxidative stress induction. The fungal vacuoles were protected against the disruptive damage even if cells were treated with H(2)O(2) alone at a lethal concentration or treated with H(2)O(2) at a sublethal concentration in combination with AmB. N-methyl-N″-dodecylguanidine was ineffective in enhancing AmB lethality or AmB-induced vacuole disruption when cells had been pretreated with ergosterol. Ergosterol-dependent mechanism is thus considered to be a possible target of N-methyl-N″-dodecylguanidine in enhancing the vacuole-targeting fungicidal activity of AmB in C. albicans cells.


Assuntos
Anfotericina B/farmacologia , Antifúngicos/farmacologia , Candida albicans/efeitos dos fármacos , Guanidinas/farmacologia , Anfotericina B/administração & dosagem , Antifúngicos/administração & dosagem , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos , Sinergismo Farmacológico , Ergosterol/farmacologia , Guanidinas/administração & dosagem , Guanidinas/química , Peróxido de Hidrogênio/administração & dosagem , Peróxido de Hidrogênio/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Vacúolos/efeitos dos fármacos
18.
AMB Express ; 1(1): 2, 2011 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-21906328

RESUMO

A bacterium Ensifer adhaerens FERM P-19486 with the ability of alliinase production was isolated from a soil sample. The enzyme was purified for characterization of its general properties and evaluation of its application in on-site production of allicin-dependent fungicidal activity. The bacterial alliinase was purified 300-fold from a cell-free extract, giving rise to a homogenous protein band on polyacrylamide gel electrophoresis. The bacterial alliinase (96 kDa) consisted of two identical subunits (48 kDa), and was most active at 60°C and at pH 8.0. The enzyme stoichiometrically converted (-)-alliin ((-)-S-allyl-L-cysteine sulfoxide) to form allicin, pyruvic acid, and ammonia more selectively than (+)-alliin, a naturally occurring substrate for plant alliinase ever known. The C-S lyase activity was also detected with this bacterial enzyme when S-alkyl-L-cysteine was used as a substrate, though such a lyase activity is absolutely absent in alliinase of plant origin. The enzyme generated a fungicidal activity against Saccharomyces cerevisiae in a time- and a dose-dependent fashion using alliin as a stable precursor. Alliinase of Ensifer adhaerens FERM P-19486 is the enzyme with a novel type of substrate specificity, and thus considered to be beneficial when used in combination with garlic enzyme with respect to absolute conversion of (±)-alliin to allicin.

19.
J Antibiot (Tokyo) ; 63(12): 689-92, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20940723

RESUMO

Allicin selectively enhances the fungicidal activity of amphotericin B (AmB). It also accelerates AmB-induced vacuole disruption but does not affect AmB-induced potassium ion efflux in Saccharomyces cerevisiae and Candida albicans. The fungicidal activity of AmB alone or combined with allicin was further evaluated based on the relationship among cell viability, vacuole disruption and potassium ion efflux in S. cerevisiae. Lethality and vacuole disruption caused by AmB alone were completely restricted when K(+) and Mg(2+) were added to the growth medium. On the other hand, in identical conditions, the combination of AmB and allicin induced both lethality and vacuole disruption. S. cerevisiae Δerg6 cells, which lack ergosterol in plasma membrane, were mostly resistant to AmB as well as the combination of AmB and allicin against both lethality and vacuole disruption. The incorporation of AmB into the cytoplasm of Δerg6 cells was significantly reduced in comparison with that in parent cells, regardless of the presence of allicin. Our results suggest that the fungicidal activity of AmB combined with allicin is involved in vacuole disruption but not in potassium ion efflux, and that the expression of allicin-mediated activity of AmB requires the presence of ergosterol in the plasma membrane.


Assuntos
Anfotericina B/farmacologia , Antifúngicos/farmacologia , Potássio/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Ácidos Sulfínicos/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Dissulfetos , Sinergismo Farmacológico , Ergosterol/metabolismo , Vacúolos/efeitos dos fármacos , Vacúolos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA