Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncol Rep ; 30(6): 2625-31, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24065141

RESUMO

The aim of the present study was to evaluate the effects of the REG Iα and REG Iß genes on lung cancer cell lines, and thereafter, the expression of REG family genes (REG Iα, REG Iß, REG III, HIP/PAP and REG IV) in lung cancer in relation to patient prognosis was evaluated. Lung adenocarcinoma (AD) and squamous cell carcinoma (SCC) cell lines expressing REG Iα or REG Iß (HLC-1 REG Iα/Iß and EBC-1 REG Iα/Iß) were established, and cell number, cell invasive activity, and anchorage-independent cell growth were compared with these variables in the control cells. The expression levels of REG family genes were evaluated by real-time RT-PCR in surgically resected lung cancers, and disease-specific survival (DSS) curves were generated. The HLC-1 REG Iα/Iß cell line showed significant increases in cell number and anchorage-independent cell growth compared with the control cells. EBC-1 REG Iα/Iß cells showed significant increases in cell invasive activity and anchorage-independent cell growth as compared with the control cells. Except for the REG Iß gene, expression of other REG family genes was observed in the surgically resected samples; however, DSS was significantly worse only in stage I patients who were positive for REG Iα expression than in patients who were negative for REG Iα expression. The effects of REG Iα on AD and SCC cells were different in the in vitro study, and a correlation between REG Iα expression and patient prognosis was noted in the in vivo study. Therefore, overexpression of REG Iα is a risk factor for poor prognosis caused by discrete mechanisms in AD and SCC patients.


Assuntos
Adenocarcinoma/genética , Carcinoma de Células Escamosas/genética , Litostatina/genética , Neoplasias Pulmonares/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Idoso , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Litostatina/biossíntese , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Estadiamento de Neoplasias , Proteínas Associadas a Pancreatite , Prognóstico
2.
J Biomed Mater Res A ; 101(10): 2838-45, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23468218

RESUMO

We evaluated the advantages of three-dimensional (3D) culture in a collagen hydrogel for stem cell differentiation, including the morphology of differentiated cells, differentiation efficiency of stem cells from aged rat and cells after passaging and freeze/thawing. Rat mesenchymal stem cells (MSCs) from young and aged rats, and MSCs after passaging and freeze/thawing were induced to differentiate into osteoblasts in 3D and 2D cultures, and histological studies were performed. Differentiation efficiency was evaluated by markers of osteoblastic differentiation including Runx2 and osterix gene expressions, osteocalcin secretion and calcium deposition. MSCs were stained positive for alkaline phosphatase in 3D and 2D cultures. However, the morphology of differentiated cells in 3D culture, which was different from that in 2D culture, was similar to that of osteoblasts in vivo. Markers of osteoblastic differentiation in MSCs from aged rats in 3D culture were higher than those in MSCs from young rats in 2D culture. Markers of osteoblastic differentiation in MSCs after passaging and freeze/thawing in 3D culture were higher than those in nonpassaged MSCs in 2D culture. These results indicate that 3D culture in a collagen hydrogel has advantages for the differentiation of MSCs into osteoblasts with a similar phenotype to that of in vivo, when using even MSCs from aged donors or after passaging and freeze/thawing.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Colágeno/farmacologia , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Células-Tronco Mesenquimais/citologia , Envelhecimento/fisiologia , Animais , Cálcio/metabolismo , Congelamento , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Ratos
3.
Eur J Appl Physiol ; 111(9): 2005-14, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21249386

RESUMO

The aim of the present study was to examine whether amount of oral antimicrobial components, human ß-defensin-2 (HBD-2), cathelicidin (LL-37), and immunoglobulin A (IgA), might be affected by prolonged strenuous exercise. Ten young male volunteers either exercised on recumbent ergometer at 75% [Formula: see text] for 60 min (exercise session) or sat quietly (resting session). Saliva samples were obtained at 60-min intervals during sessions for measurements of saliva antimicrobial components (HBD-2, LL-37, and IgA), saliva cortisol and osmolality. Saliva flow rate was decreased and saliva osmolality was increased during the 60-min exercise. Saliva HBD-2 and LL-37 concentrations and secretion rates were increased during and after the exercise, whereas saliva IgA concentration and secretion rates were decreased after the exercise. Saliva cortisol was increased during and after the exercise. The areas under the curve of the time courses of saliva levels of HBD-2 and LL-37 were negatively correlated with those of cortisol levels in saliva. The present findings suggested that a single bout of prolonged strenuous exercise caused a transient increase in the oral HBD-2 and LL-37 levels.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Exercício Físico/fisiologia , Hidrocortisona/metabolismo , Imunoglobulina A/metabolismo , Esforço Físico/fisiologia , Saliva/metabolismo , Adulto , Algoritmos , Peptídeos Catiônicos Antimicrobianos/análise , Teste de Esforço , Humanos , Hidrocortisona/análise , Imunoglobulina A/análise , Masculino , Saliva/química , Proteínas e Peptídeos Salivares/análise , Proteínas e Peptídeos Salivares/metabolismo , Fatores de Tempo , Adulto Jovem
4.
Microbiol Immunol ; 55(3): 141-53, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21204951

RESUMO

The oral anaerobic bacterium Porphyromonas gingivalis, which is implicated as an important pathogen for chronic periodontitis, requires heme for its growth. Non-iron metalloporphyrins, In-PPIX and Ga-PPIX, were examined for antibacterial effects on P. gingivalis. Both In-PPIX and Ga-PPIX caused retardation of P. gingivalis growth in a dose-dependent fashion. Microarray and qPCR analyses revealed that In-PPIX treatment upregulated the expression of several genes encoding proteins including ClpB and ClpC, which are members of the Clp (caseinolytic protease, Hsp100) family, and aRNR, aRNR-activating protein and thioredoxin reductase, whereas In-PPIX treatment had no effect on the expression of genes encoding proteins involved in heme uptake pathways, Hmu-mediated, Iht-mediated and Tlr-mediated pathways. P. gingivalis ihtA and ihtB mutants were more resistant to In-PPIX than was the wild-type parent, whereas hmuR and tlr mutants did not show such resistance to In-PPIX. The results suggest that In-PPIX is incorporated by the Iht-mediated heme uptake pathway and that it influences protein quality control and nucleotide metabolism and retards growth of P. gingivalis.


Assuntos
Antibacterianos/farmacologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Metaloporfirinas/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , Antibacterianos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Perfilação da Expressão Gênica , Heme/metabolismo , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Mutação , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/crescimento & desenvolvimento
5.
Infect Immun ; 78(6): 2846-56, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20351137

RESUMO

Porphyromonas gingivalis is one of the most etiologically important microorganisms in periodontal disease. We found in a previous study that PG1385 (TprA) protein, a tetratricopeptide repeat (TPR) protein, was upregulated in P. gingivalis wild-type cells placed in a mouse subcutaneous chamber and that a tprA mutant was clearly less virulent in the mouse subcutaneous abscess model (M. Yoshimura et al., Oral Microbiol. Immunol. 23:413-418, 2008). In the present study, we investigated the gene expression profile of tprA mutant cells placed in a mouse subcutaneous chamber and found that 9 genes, including PG2102 (tapA), PG2101 (tapB), and PG2100 (tapC) genes, were downregulated in the tprA mutant compared with those in the wild type. Expression of a cluster of tapA, tapB, and tapC genes of the mutant was also downregulated in an in vitro culture with enriched brain heart infusion medium. The TprA protein has three TPR motifs known as a protein-protein interaction module. Yeast two-hybrid system analysis and in vitro protein binding assays with immunoprecipitation and surface plasmon resonance detection revealed that the TprA protein could bind to TapA and TapB proteins. TprA and TapB proteins were located in the periplasmic space, whereas TapA, which appeared to be one of the C-terminal domain family proteins, was located at the outer membrane. We constructed tapA, tapB, and tapC single mutants and a tapA-tapB-tapC deletion mutant. In the mouse subcutaneous infection experiment, all of the mutants were less virulent than the wild type. These results suggest that TprA, TapA, TapB, and TapC are cooperatively involved in P. gingivalis virulence.


Assuntos
Proteínas de Bactérias/metabolismo , Porphyromonas gingivalis/patogenicidade , Fatores de Virulência/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/genética , Infecções por Bacteroidaceae/microbiologia , Meios de Cultura/química , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Imunoprecipitação , Camundongos , Camundongos Endogâmicos BALB C , Periplasma/química , Ligação Proteica , Mapeamento de Interação de Proteínas , Infecções dos Tecidos Moles/microbiologia , Ressonância de Plasmônio de Superfície , Técnicas do Sistema de Duplo-Híbrido , Virulência , Fatores de Virulência/genética
6.
J Periodontol ; 80(11): 1845-51, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19905954

RESUMO

BACKGROUND: Hydrogen sulfide is one of the predominant volatile sulfur compounds (VSCs) produced by oral bacteria. This study developed and evaluated a system for detecting hydrogen sulfide production by oral bacteria. METHODS: L-methionine-alpha-deamino-gamma-mercaptomethane-lyase (METase) and beta carbon-sulfur (beta C-S) lyase were used to degrade homocysteine and cysteine, respectively, to produce hydrogen sulfide. Enzymatic reactions resulting in hydrogen sulfide production were assayed by reaction with bismuth trichloride, which forms a black precipitate when mixed with hydrogen sulfide. The enzymatic activities of various oral bacteria that result in hydrogen sulfide production and the capacity of bacteria from periodontal sites to form hydrogen sulfide in reaction mixtures containing L-cysteine or DL-homocysteine were assayed. RESULTS: With L-cysteine as the substrate, Streptococcus anginosus FW73 produced the most hydrogen sulfide, whereas Porphyromonas gingivalis American Type Culture Collection (ATCC) 33277 and W83 and Fusobacterium nucleatum ATCC 10953 produced approximately 35% of the amount produced by the P. gingivalis strains. Finally, the hydrogen sulfide found in subgingival plaque was analyzed. Using bismuth trichloride, the hydrogen sulfide produced by oral bacteria was visually detectable as a black precipitate. CONCLUSIONS: Hydrogen sulfide production by oral bacteria was easily analyzed using bismuth trichloride. However, further innovation is required for practical use.


Assuntos
Bactérias/metabolismo , Cisteína/metabolismo , Homocisteína/metabolismo , Sulfeto de Hidrogênio/metabolismo , Boca/microbiologia , Doenças Periodontais/microbiologia , Adulto , Idoso , Bacteroides/metabolismo , Bismuto , Liases de Carbono-Enxofre/metabolismo , Cloretos , Placa Dentária/microbiologia , Enterococcus/metabolismo , Feminino , Fusobacterium nucleatum/metabolismo , Humanos , Indicadores e Reagentes , Masculino , Pessoa de Meia-Idade , Bolsa Periodontal/microbiologia , Porphyromonas gingivalis/metabolismo , Prevotella intermedia/metabolismo , Prevotella nigrescens/metabolismo , Streptococcus/metabolismo , Streptococcus anginosus/metabolismo
7.
PLoS Pathog ; 5(10): e1000643, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19876387

RESUMO

In spite of the importance of hyaluronan in host protection against infectious organisms in the alveolar spaces, its role in mycobacterial infection is unknown. In a previous study, we found that mycobacteria interact with hyaluronan on lung epithelial cells. Here, we have analyzed the role of hyaluronan after mycobacterial infection was established and found that pathogenic mycobacteria can grow by utilizing hyaluronan as a carbon source. Both mouse and human possess 3 kinds of hyaluronan synthases (HAS), designated HAS1, HAS2, and HAS3. Utilizing individual HAS-transfected cells, we show that HAS1 and HAS3 but not HAS2 support growth of mycobacteria. We found that the major hyaluronan synthase expressed in the lung is HAS1, and that its expression was increased after infection with Mycobacterium tuberculosis. Histochemical analysis demonstrated that hyaluronan profoundly accumulated in the granulomatous legion of the lungs in M. tuberculosis-infected mice and rhesus monkeys that died from tuberculosis. We detected hyaluronidase activity in the lysate of mycobacteria and showed that it was critical for hyaluronan-dependent extracellular growth. Finally, we showed that L-Ascorbic acid 6-hexadecanoate, a hyaluronidase inhibitor, suppressed growth of mycobacteria in vivo. Taken together, our data show that pathogenic mycobacteria exploit an intrinsic host-protective molecule, hyaluronan, to grow in the respiratory tract and demonstrate the potential usefulness of hyaluronidase inhibitors against mycobacterial diseases.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Ácido Hialurônico/metabolismo , Mycobacterium tuberculosis/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Contagem de Colônia Microbiana , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Glicosaminoglicanos/farmacologia , Histocitoquímica , Humanos , Hialuronan Sintases , Ácido Hialurônico/farmacologia , Pulmão/química , Pulmão/metabolismo , Pulmão/microbiologia , Macaca mulatta , Masculino , Camundongos , Mycobacterium bovis/fisiologia , Mycobacterium tuberculosis/metabolismo , Ratos
8.
Jpn J Infect Dis ; 62(3): 182-6, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19468176

RESUMO

Medical treatment of pulmonary Mycobacterium avium complex (MAC) disease does not always provide curative effects and is frequently hampered by recurrence. This suggests the presence of a reservoir for MAC in the environment surrounding patients. We previously reported the recovery of MAC isolates from the residential bathrooms of outpatients. In the present study, to ascertain the colonizing sites and the possibility of an MAC reservoir in the bathrooms of patients, we tested the recovery and the genetic diversity of MAC isolates from 6 sites of specimens, including 2 additional sampling sites, inside the showerhead and the bathtub inlet, in the residential bathrooms of patients with pulmonary MAC disease. MAC isolates were recovered from 15 out of the 29 bathrooms (52%), including specimens from 14 bathtub inlets and 3 showerheads. Nearly half of these bathrooms (7/15) contained MAC strains that were identical or similar to their respective clinical isolates Additionally, in 5 out of 15 bathrooms, polyclonal colonization was revealed by pulsed-field gel electrophoresis. The results imply that colonization of MAC organisms in the bathrooms of MAC patients occurs predominantly in the bathtub inlets, and there is thus a risk of infection and/or reinfection for patients via use of the bathtub and other sites in the bathroom.


Assuntos
Banhos , Contaminação de Equipamentos , Hospitais , Complexo Mycobacterium avium/isolamento & purificação , Quartos de Pacientes , Biofilmes , Distribuição de Qui-Quadrado , Eletroforese em Gel de Campo Pulsado , Zeladoria Hospitalar , Humanos , Japão , Complexo Mycobacterium avium/genética , Complexo Mycobacterium avium/crescimento & desenvolvimento , Infecção por Mycobacterium avium-intracellulare/microbiologia , Engenharia Sanitária , Estatísticas não Paramétricas , Microbiologia da Água
9.
Microb Pathog ; 46(1): 6-12, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19013228

RESUMO

Mycobacterium avium complex (MAC) disease has been increasing worldwide not only in immunocompromised but also in immunocompetent humans. However, the relationship between mycobacterial strain virulence and disease progression in immunocompetent humans is unclear. In this study, we isolated 6 strains from patients with pulmonary MAC disease. To explore the virulence, we examined the growth in human THP-1 macrophages and pathogenicity in C57BL/6 mice. We found that one strain, designated 198, which was isolated from a patient showing the most progressive disease, persisted in THP-1 cells. In addition, strain 198 grew to a high bacterial load with strong inflammation in mouse lungs and spleens 16 weeks after infection. To our knowledge, strain 198 is the first isolated MAC strain that exhibits hypervirulence consistently for the human patient, human macrophages in vitro, and even for immunocompetent mice. Other strains showed limited survival and weak virulence both in macrophages and in mice, uncorrelated to disease progression in human patients. We demonstrated that there is a hypervirulent clinical MAC strain whose experimental virulence corresponds to the serious disease progression in the patients. The existence of such strain suggests the involvement of bacterial virulence in the pathogenesis of pulmonary MAC disease in immunocompetent status.


Assuntos
Imunocompetência , Complexo Mycobacterium avium/isolamento & purificação , Complexo Mycobacterium avium/patogenicidade , Infecção por Mycobacterium avium-intracellulare/patologia , Tuberculose Pulmonar/patologia , Animais , Linhagem Celular , Feminino , Humanos , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/citologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Monócitos/microbiologia , Complexo Mycobacterium avium/crescimento & desenvolvimento , Infecção por Mycobacterium avium-intracellulare/microbiologia , Tuberculose Pulmonar/microbiologia , Virulência
10.
J Bacteriol ; 189(22): 8241-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17873049

RESUMO

Bacteria coordinate assembly of the cell wall as well as synthesis of cellular components depending on the growth state. The mycobacterial cell wall is dominated by mycolic acids covalently linked to sugars, such as trehalose and arabinose, and is critical for pathogenesis of mycobacteria. Transfer of mycolic acids to sugars is necessary for cell wall biogenesis and is mediated by mycolyltransferases, which have been previously identified as three antigen 85 (Ag85) complex proteins. However, the regulation mechanism which links cell wall biogenesis and the growth state has not been elucidated. Here we found that a histone-like protein has a dual concentration-dependent regulatory effect on mycolyltransferase functions of the Ag85 complex through direct binding to both the Ag85 complex and the substrate, trehalose-6-monomycolate, in the cell wall. A histone-like protein-deficient Mycobacterium smegmatis strain has an unusual crenellated cell wall structure and exhibits impaired cessation of glycolipid biosynthesis in the growth-retarded phase. Furthermore, we found that artificial alteration of the amount of the extracellular histone-like protein and the Ag85 complex changes the growth rate of mycobacteria, perhaps due to impaired down-regulation of glycolipid biosynthesis. Our results demonstrate novel regulation of cell wall assembly which has an impact on bacterial growth.


Assuntos
Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Mycobacterium smegmatis/citologia , Mycobacterium smegmatis/metabolismo , Aciltransferases/metabolismo , Adesinas Bacterianas/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Fatores Corda , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica , Glicolipídeos , Histonas/química , Histonas/genética , Mycobacterium bovis , Mycobacterium smegmatis/genética , Ligação Proteica
11.
Infect Immun ; 74(5): 2544-51, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16622189

RESUMO

Extracellular proteinaceous factors of Porphyromonas gingivalis, a periodontal pathogen, that influence receptor activator of nuclear factor-kappaB (NF-kappaB) ligand (RANKL)-induced osteoclastogenesis from bone marrow macrophages were investigated. The culture supernatant of P. gingivalis had the ability to inhibit RANKL-induced in vitro osteoclastogenesis. A major protein of the culture supernatant, hemoglobin receptor protein (HbR), suppressed RANKL-induced osteoclastogenesis in a dose-dependent fashion. HbR markedly inhibited RANKL-induced osteoclastogenesis when present in the culture for the first 24 h after addition of RANKL, whereas no significant inhibition was observed when HbR was added after 24 h or later, implying that HbR might interfere with only the initial stage of RANKL-mediated differentiation. HbR tightly bound to bone marrow macrophages and had the ability to induce phosphorylation of ERK, p38, NF-kappaB, and Akt. RANKL-induced phosphorylation of ERK, p38, and NF-kappaB was not suppressed by HbR, but that of Akt was markedly suppressed. HbR inhibited RANKL-mediated induction of c-Fos and NFATc1. HbR could induce beta interferon (IFN-beta) from bone marrow macrophages, but the induction level of IFN-beta might not be sufficient to suppress RANKL-mediated osteoclastogenesis, implying presence of an IFN-beta-independent pathway in HbR-mediated inhibition of osteoclastogenesis. Since rapid and extensive destruction of the alveolar bone causes tooth loss, resulting in loss of the gingival crevice that is an anatomical niche for periodontal pathogens such as P. gingivalis, the suppressive effect of HbR on osteoclastogenesis may help the microorganism exist long in the niche.


Assuntos
Proteínas de Bactérias/fisiologia , Células da Medula Óssea/citologia , Proteínas de Transporte/antagonistas & inibidores , Macrófagos/citologia , Glicoproteínas de Membrana/antagonistas & inibidores , Osteoclastos/fisiologia , Osteogênese , Porphyromonas gingivalis/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Apoptose , Diferenciação Celular , Interferon beta/biossíntese , Masculino , Camundongos , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Transdução de Sinais
12.
Biosci Biotechnol Biochem ; 69(11): 2244-6, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16306712

RESUMO

We have developed a new enzymatic assay for determining L-cysteine concentration. The method involves the use of betaC-S lyase from Streptococcus anginosus, which catalyzes the alpha,beta-elimination of L-cysteine to hydrogen sulfide, pyruvate, and ammonia. The production of pyruvate is measured by D-lactate dehydrogenase and NADH. The decrease in NADH was proportional to the L-cysteine concentration up to 1.0 mM. When serum samples were used, within-day and day-to-day coefficient variations were below 4%. This method is simple, and can easily and reliably be used for accurate determination of L-cysteine concentration in serum or other samples.


Assuntos
Cistationina gama-Liase/metabolismo , Cisteína/análise , Calibragem , Cisteína/sangue , Cisteína/metabolismo , Humanos , L-Lactato Desidrogenase/metabolismo , NAD/análise , NAD/metabolismo , Ácido Pirúvico/análise , Ácido Pirúvico/metabolismo , Reprodutibilidade dos Testes , Streptococcus anginosus/enzimologia
13.
Microbiology (Reading) ; 151(Pt 3): 841-853, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15758230

RESUMO

Porphyromonas gingivalis, an obligately anaerobic bacterium, is implicated as a major pathogen in the development and progression of chronic periodontitis. Although expression of several virulence factors of the bacterium has been found to be affected by environmental stress such as entrance into the stationary growth phase and heat, there is relatively little information on the mechanisms that may operate in the bacterium in response to environmental stress. In this study, a novel protein (UstA) was investigated that was initially identified following two-dimensional gel analysis. Expression of UstA was upregulated in stationary phase or by exposure to atmospheric oxygen. N-terminal sequencing and database analysis with the P. gingivalis genome sequence revealed that the UstA-encoding gene (ustA) was located upstream of a homologue of the usp gene encoding the universal stress protein on the chromosome. The ustA gene appeared to be transcribed in a monocistronic fashion, as revealed by primer extension and Northern blot analysis. To elucidate the role of UstA in the bacterium, chromosomal mutants carrying a disruption of the ustA gene were constructed. The ustA mutant grew slower than the wild-type parent strain in rich medium, resulting in a lower yield in stationary phase. Furthermore, in this mutant, expression levels of the P. gingivalis homologues of superoxide dismutase, thiol peroxidase and thioredoxin were markedly higher than those in the wild-type, especially in stationary phase. The ustA mutant was more resistant to diamide, a thiol-specific oxidant, than the wild-type. In addition, the ustA mutation suppressed hypersensitivities of the oxyR mutant to diamide, metronidazole and mitomycin C. These results suggest that UstA may play a significant role in oxidative stress responses in the bacterium.


Assuntos
Proteínas de Bactérias , Regulação Bacteriana da Expressão Gênica , Porphyromonas gingivalis/crescimento & desenvolvimento , Regulação para Cima , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Meios de Cultura , Eletroforese em Gel Bidimensional , Humanos , Dados de Sequência Molecular , Estresse Oxidativo , Peroxidases/metabolismo , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/metabolismo , Compostos de Sulfidrila/metabolismo , Superóxido Dismutase/metabolismo , Tiorredoxinas/metabolismo
14.
Microbiology (Reading) ; 148(Pt 12): 3961-3970, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12480900

RESUMO

Hydrogen sulfide is highly toxic to mammalian cells. It has also been postulated that hydrogen sulfide modifies haemoglobin resulting in haemolysis. The enzyme that produces hydrogen sulfide from L-cysteine was purified from Streptococcus anginosus. Using the N-terminal amino acid sequence of the purified enzyme, the lcd gene encoding L-cysteine desulfhydrase was cloned; the recombinant protein was then purified to examine its enzymic and biological characteristics. This L-cysteine desulfhydrase had the Michaelis-Menten kinetics K(m)=0.62 mM and V(max)=163 micro mol min(-1) mg(-1). DL-Cystathionine, L-cystine, S-(2-aminoethyl)-L-cysteine, 3-chloro-DL-alanine and S-methyl-L-cysteine were substrates for the enzyme, whereas D-cysteine, DL-homocysteine, L-methionine, DL-serine, DL-alanine, L-cysteine methyl ester, L-tryptophan, L-tyrosine and L-phenylalanine were not. These findings suggest that this L-cysteine desulfhydrase is a C-S lyase that catalyses the alpha,beta-elimination (alphaC-N and betaC-S) reaction. In addition, it is demonstrated that the hydrogen sulfide produced by this enzyme caused the modification and release of haemoglobin in sheep erythrocytes.


Assuntos
Cistationina gama-Liase/genética , Cistationina gama-Liase/metabolismo , Liases/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cistationina gama-Liase/química , Cisteína/metabolismo , Hemoglobinas/metabolismo , Humanos , Sulfeto de Hidrogênio/metabolismo , Liases/química , Liases/genética , Dados de Sequência Molecular , Análise de Sequência de DNA , Especificidade por Substrato
15.
FEMS Microbiol Lett ; 215(1): 75-80, 2002 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-12393204

RESUMO

Hydrogen sulfide and methyl mercaptan are the two major compounds associated with oral malodor. These compounds are highly toxic, and are thought to play an important role in periodontal disease. Fusobacterium nucleatum, an oral bacterium, produces large amounts of hydrogen sulfide from L-cysteine by the enzymatic action of L-cysteine desulfhydrase. We cloned and sequenced the cdl gene encoding L-cysteine desulfhydrase from F. nucleatum ATCC 10953, and revealed that the structural cdl gene consists of 921 bp and encodes a 33.4-kDa protein. The cloned gene was inserted into an expression vector, pDEST17, and expressed in Escherichia coli as a fused protein. The purified enzyme was tested for substrate specificity using various SH-containing compounds. Only L-cysteine served as a substrate for L-cysteine desulfhydrase to produce hydrogen sulfide.


Assuntos
Cistationina gama-Liase/genética , Cistationina gama-Liase/metabolismo , Fusobacterium nucleatum/enzimologia , Fusobacterium nucleatum/genética , Sequência de Aminoácidos , Clonagem Molecular , Cistationina gama-Liase/isolamento & purificação , Halitose/microbiologia , Humanos , Sulfeto de Hidrogênio/metabolismo , Dados de Sequência Molecular , Doenças Periodontais/microbiologia
16.
FEBS Lett ; 523(1-3): 119-22, 2002 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-12123816

RESUMO

The antibacterial agent 3-chloro-DL-alanine (3CA) is an inhibitor of peptidoglycan synthesis. Fusobacterium nucleatum and Porphyromonas gingivalis, the bacteria responsible for oral malodor, are shown to be resistant to 1 mM 3CA, whereas Streptococcus mutans and Escherichia coli are sensitive to this antibacterial agent at the same concentration. We isolated the 3CA resistance gene from F. nucleatum and showed that the gene encodes an L-methionine-alpha-deamino-gamma-mercaptomethane-lyase that catalyzes the alpha,gamma-elimination of L-methionine to produce methyl mercaptan. The enzyme also exhibits 3CA chloride-lyase (deaminating) activity. This antibacterial agent is expected to be useful for specific selection of malodorous oral bacteria producing high amounts of methyl mercaptan.


Assuntos
Liases de Carbono-Enxofre/metabolismo , Farmacorresistência Bacteriana/genética , Fusobacterium nucleatum/enzimologia , Compostos de Sulfidrila/metabolismo , beta-Alanina/análogos & derivados , beta-Alanina/farmacologia , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Liases de Carbono-Enxofre/genética , Desaminação , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/genética , Metionina/metabolismo , Testes de Sensibilidade Microbiana , Porphyromonas gingivalis/efeitos dos fármacos , Especificidade da Espécie , beta-Alanina/metabolismo
17.
Microbes Infect ; 4(6): 679-83, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12048037

RESUMO

Volatile sulfur compounds (VSCs), including hydrogen sulfide, methyl mercaptan, and dimethyl sulfide, are primarily responsible for oral malodor. Recently, the mgl gene encoding L-methionine-alpha-deamino-gamma-mercaptomethane-lyase, which produces methyl mercaptan, was cloned from Porphyromonas gingivalis. This article discusses the mechanism and pathogenic role of the formation of VSCs by oral bacteria.


Assuntos
Infecções por Bacteroidaceae/complicações , Halitose/microbiologia , Boca/microbiologia , Porphyromonas gingivalis/metabolismo , Adolescente , Adulto , Infecções por Bacteroidaceae/metabolismo , Humanos , Pessoa de Meia-Idade , Compostos de Sulfidrila/metabolismo
18.
Biochem Biophys Res Commun ; 292(4): 964-8, 2002 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-11944909

RESUMO

Methyl mercaptan is a major component responsible for oral malodor. This compound arises from the bacterial metabolism of methionine. Here we show that the growth of Porphyromonas gingivalis, a periodontal microorganism that produces large amounts of methyl mercaptan, was strongly inhibited by l-trifluoromethionine (TFM), a fluorinated derivative of methionine. In contrast, TFM had no effect on the growth of bacteria which do not produce methyl mercaptan. In addition, the survival rate of P. gingivalis-infected mice was remarkably increased by the co-injection of TFM. These results suggest that TFM is a promising antibacterial agent specific to the malodorous oral bacteria.


Assuntos
Infecções por Bacteroidaceae/tratamento farmacológico , Liases de Carbono-Enxofre/metabolismo , Metionina/análogos & derivados , Metionina/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento , Animais , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/patologia , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Feminino , Halitose/etiologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Porphyromonas gingivalis/metabolismo , Porphyromonas gingivalis/patogenicidade , Compostos de Sulfidrila/metabolismo , Taxa de Sobrevida , Virulência/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...