Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Curr Microbiol ; 81(8): 243, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38935166

RESUMO

Clostridium perfringens is one of the critical causative agents causing diarrhea in piglets, with significant economic losses to the pig industry. Under normal gut microbiota homeostasis and well-managed barns, diarrhea caused by C. perfringens could be controlled. Some reports show that probiotics, such as Bacillus subtilis, are beneficial in preventing necrotic enteritis (NE) in chickens, but few reports on piglets. Clostridium perfringens was found in the piglets' diarrhea with intestinal microbiota dysbiosis in our survey. Bacillus subtilis G2B9-Q, which was isolated from the feces of healthy pigs, was found to have anti-Clostridium activity after screening. Clostridium perfringens was used to challenge mice by intraperitoneal injection for modeling to evaluate the anti-infective activity of cell-free supernatant (CFS) of B. subtilis G2B9-Q and different concentrations of B. subtilis G2B9-Q by oral administration. The results showed that G2B9-Q can mitigate intestinal lesions caused by C. perfringens infection, reduce inflammatory reactions, and modulate intestinal microbiota. The CFS of G2B9-Q can alleviate the pathological damage of intestinal tissues caused by C. perfringens infection, reduce the concentration of TNF-α and IL-10 in the sera of mice, as well as the relative expression levels of alpha toxin (CPA), perfringolysin O (PFO) toxin, IL-10, IL-22, and TNF-α in the jejunum and colon tissues, and alleviate the changes in gut microbiota structure caused by C. perfringens infection, which showed better therapeutic effects and indicated that the metabolites of G2B9-Q are essential mediators for their beneficial effects. Therefore, the CFS of G2B9-Q could potentially replace antibiotics in treating C. perfringens infection.


Assuntos
Bacillus subtilis , Infecções por Clostridium , Clostridium perfringens , Microbioma Gastrointestinal , Probióticos , Animais , Infecções por Clostridium/imunologia , Infecções por Clostridium/microbiologia , Bacillus subtilis/genética , Clostridium perfringens/imunologia , Camundongos , Probióticos/administração & dosagem , Microbioma Gastrointestinal/efeitos dos fármacos , Intestinos/microbiologia , Intestinos/imunologia , Suínos , Diarreia/microbiologia , Diarreia/imunologia , Fezes/microbiologia , Modelos Animais de Doenças
2.
J Dairy Sci ; 106(12): 9174-9185, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37641240

RESUMO

Bovine mastitis is the most common and costly disease affecting dairy cattle throughout the world. Enterococcus faecalis is one of the environmental origin mastitis-causing pathogens. The treatment of bovine mastitis is primarily based on antibiotics. Due to the negative impact of developing antibiotic resistance and adverse effects on soil and water environments, the trend toward use of nonantibiotic treatments is increasing. Phages may represent a promising alternative treatment strategy. However, it is unknown whether phages have therapeutic effects on E. faecalis-induced mastitis. Thus, the objective of this study was to investigate the degree of protection conferred by a phage during murine mastitis caused by multidrug-resistant E. faecalis. Enterococcus faecalis was isolated from the milk of dairy cows with mastitis, and a phage was isolated using the E. faecalis isolates as hosts. The bactericidal ability of the phage against E. faecalis and the ability to prevent biofilm formation were determined in vitro. The therapeutic potential of the phage on murine mastitis was evaluated in vivo. We isolated 14 strains of E. faecalis from the milk of cows with mastitis, all of which exhibited multidrug resistance, and most (10/14) could form strong biofilms. Subsequently, a new phage (EF-N13) was isolated using the multidrug-resistant E. faecalis N13 (isolated from mastitic milk) as the host. The phage EF-N13 belongs to the family Myoviridae, which has short latent periods (5 min) and high bursts (284 pfu/cell). The genome of EF-N13 lacked bacterial virulence-, antibiotic resistance-, and lysogenesis-related genes. Furthermore, bacterial loading in the raw milk medium was significantly reduced by EF-N13 and was unaffected by potential IgG antibodies. In fact, EF-N13 could effectively prevent the formation of biofilm by multidrug-resistant E. faecalis. All of these characteristics suggest that EF-N13 has potential as mastitis therapy. In vivo, 1 × 105 cfu/gland of multidrug-resistant E. faecalis N13 resulted in mastitis development within 24 h. A single dose of phage EF-N13 (1 × 104, 1 × 105, or 1 × 106 pfu/gland) could significantly decrease bacterial counts in the mammary gland at 24 h postinfection. Histopathological observations demonstrated that treatment with phage EF-N13 effectively alleviated mammary gland inflammation and damage. This effect was confirmed by the lower levels of proinflammatory cytokines IL-6, IL-1ß, and tumor necrosis factor-α in the mammary gland treated with phage EF-N13 compared with those treated with phosphate-buffered saline. Overall, the data underscored the potential of phage EF-N13 as an alternative therapy for bovine mastitis caused by multidrug-resistant E. faecalis.


Assuntos
Bacteriófagos , Doenças dos Bovinos , Mastite Bovina , Animais , Bovinos , Feminino , Camundongos , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Bacteriófagos/genética , Enterococcus faecalis , Mastite Bovina/terapia , Mastite Bovina/microbiologia
3.
BMC Cancer ; 22(1): 864, 2022 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-35941558

RESUMO

BACKGROUND: IFN-γ has been traditionally recognized as an inflammatory cytokine that involves in inflammation and autoimmune diseases. Previously we have shown that sustained IFN-γ induced malignant transformation of bovine mammary epithelial cells (BMECs) via arginine depletion. However, the molecular mechanism underlying this is still unknown. METHODS: In this study, the amino acids contents in BMECs were quantified by a targeted metabolomics method. The acquisition of differentially expressed genes was mined from RNA-seq dataset and analyzed bioinformatically. Quantitative reverse transcription polymerase chain reaction (qRT-PCR), enzyme-linked immunosorbent assay (ELISA), western blotting, and immunohistochemistry (IHC) assay were performed to detect gene mRNA and protein expression levels. CCK-8 and would healing assays were used to detect cell proliferation and migration abilities, respectively. Cell cycle phase alternations were analyzed by flow cytometry. RESULTS: The targeted metabolomics analysis specifically discovered IFN-γ induced arginine depletion through accelerating arginine catabolism and inhibiting arginine anabolism in BMECs. Transcriptome analysis identified leucine aminopeptidase 3 (LAP3), which was regulated by p38 and ERK MAPKs, to downregulate arginine level through interfering with argininosuccinate synthetase (ASS1) as IFN-γ stimulated. Moreover, LAP3 also contributed to IFN-γ-induced malignant transformation of BMECs by upregulation of HDAC2 (histone deacetylase 2) expression and promotion of cell cycle proteins cyclin A1 and D1 expressions. Arginine supplementation did not affect LAP3 and HDAC2 expressions, but slowed down cell cycle process of malignant BMECs. In clinical samples of patients with breast cancer, LAP3 was confirmed to be upregulated, while ASS1 was downregulated compared with healthy control. CONCLUSIONS: These results demonstrated that LAP3 mediated IFN-γ-induced arginine depletion to malignant transformation of BMECs. Our findings provide a potential therapeutic target for breast cancer both in humans and dairy cows.


Assuntos
Arginina , Neoplasias da Mama , Leucil Aminopeptidase/metabolismo , Animais , Arginina/metabolismo , Argininossuccinato Sintase/metabolismo , Mama/metabolismo , Neoplasias da Mama/metabolismo , Bovinos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Interferon gama/metabolismo
4.
Vet Res ; 52(1): 30, 2021 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-33618766

RESUMO

Host proteins interacting with pathogens are receiving more attention as potential therapeutic targets in molecular medicine. Streptococcus suis serotype 2 (SS2) is an important cause of meningitis in both humans and pigs worldwide. SS2 Enolase (Eno) has previously been identified as a virulence factor with a role in altering blood brain barrier (BBB) integrity, but the host cell membrane receptor of Eno and The mechanism(s) involved are unclear. This study identified that SS2 Eno binds to 40S ribosomal protein SA (RPSA) on the surface of porcine brain microvascular endothelial cells leading to activation of intracellular p38/ERK-eIF4E signalling, which promotes intracellular expression of HSPD1 (heat-shock protein family D member 1), and initiation of host-cell apoptosis, and increased BBB permeability facilitating bacterial invasion. This study reveals novel functions for the host-interactional molecules RPSA and HSPD1 in BBB integrity, and provides insight for new therapeutic strategies in meningitis.


Assuntos
Barreira Hematoencefálica , Células Endoteliais/metabolismo , Fosfopiruvato Hidratase/metabolismo , Proteínas Ribossômicas/metabolismo , Infecções Estreptocócicas/veterinária , Streptococcus suis/metabolismo , Animais , Apoptose , Técnicas de Cocultura , Células Endoteliais/microbiologia , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Camundongos , Ligação Proteica , Sorogrupo , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Streptococcus suis/patogenicidade , Suínos , Doenças dos Suínos/microbiologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Appl Environ Microbiol ; 86(22)2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-32887718

RESUMO

Salmonella enterica subsp. enterica serovar Abortusequi is a frequently reported pathogen causing abortion in mares. In this study, the preventive and therapeutic effects of phage PIZ SAE-01E2 against S Abortusequi in a mouse model of abortion were investigated. Phage PIZ SAE-01E2 was stable at different temperatures (4 to 70°C) and pH values (pH 4 to 10) and could lyse the majority of the Salmonella serogroup O:4 and O:9 strains tested (25/28). There was no lysogeny-related, toxin, or antibiotic resistance-related gene in the genome of PIZ SAE-01E2. All of these characteristics indicate that PIZ SAE-01E2 has the potential for use in phage therapy. In in vivo experiments, 2 × 103 CFU/mouse of S Abortusequi ATCC 9842 was sufficient to lead to murine abortion (gestational day 14.5) within 48 h. A single intraperitoneal inoculation of PIZ SAE-01E2 (108 PFU/mouse, multiplicity of infection = 105) 1 h before or after S Abortusequi challenge provided effective protection to all pregnant mice (10/10). After 24 h of treatment with phage PIZ SAE-01E2, the bacterial loads in both the placenta and the uterus of the infected mice were significantly decreased (<102 CFU/g) compared to those in the placenta and the uterus of the mice in the control group (>106 CFU/g). In addition, the levels of inflammatory cytokines in the placenta and blood of the mice in the phage administration groups were significantly reduced (P < 0.05) compared to those in the placenta and blood of the mice in the control group. Altogether, these findings indicate that PIZ SAE-01E2 shows the potential to block abortions induced by S Abortusequi in vivoIMPORTANCES Abortusequi is an important pathogen that can induce abortions in mares. Although S Abortusequi has been well controlled in Europe and the United States due to strict breeding and health policies, it is still widespread in African and Asian countries and has proven difficult to control. In China, abortions caused by S Abortusequi have also been reported in donkeys. So far, there is no commercial vaccine. Thus, exploiting alternative efficient and safe strategies to control S Abortusequi infection is essential. In this study, a new lytic phage, PIZ SAE-01E2, infecting S Abortusequi was isolated, and the characteristics of PIZ SAE-01E2 indicated that it has the potential for use in phage therapy. A single intraperitoneal inoculation of PIZ SAE-01E2 before or after S Abortusequi challenge provided effective protection to all pregnant mice. Thus, PIZ SAE-01E2 showed the potential to block abortions induced by S Abortusequi in vivo.


Assuntos
Aborto Animal/prevenção & controle , Bacteriófagos/fisiologia , Doenças dos Cavalos/prevenção & controle , Salmonelose Animal/prevenção & controle , Salmonella/fisiologia , Aborto Animal/microbiologia , Aborto Animal/virologia , Animais , Feminino , Doenças dos Cavalos/microbiologia , Doenças dos Cavalos/virologia , Cavalos , Camundongos , Camundongos Endogâmicos ICR , Gravidez , Salmonelose Animal/microbiologia , Salmonelose Animal/virologia
6.
Anticancer Drugs ; 31(4): 403-410, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31917701

RESUMO

Our retrospective study assessed the efficacy and safety of irinotecan plus raltitrexed in esophageal squamous cell cancer (ESCC) patients who were previously treated with multiple systemic therapies. Between January 2016 and December 2018, records of 38 ESCC patients who underwent irinotecan plus raltitrexed chemotherapy after at least one line of chemotherapy were reviewed. Efficacy assessment was performed every two cycles according to the RECIST version 1.1. A total of 95 cycles of chemotherapy were administered, and the median course was 3 (range 2-6). There was no treatment-related death. Nine patients had partial response, 21 had stable disease and eight had progressive disease. The overall objective response rate was 23.68% (9/38) and the disease control rate was78.94% (30/38). After a median follow-up of 18.5 months, the median progression-free survival and overall survival were 105 and 221 days, respectively. There were five patients (13.15%) with grade 3/4 leukopenia, three patients (7.89%) with grade 3/4 neutropenia and one patient (2.63%) with grade 3/4 diarrhea. The combination of irinotecan plus raltitrexed was effective for pretreated ESCC patients. Further studies are needed to determine the optimal dose of the two drugs.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Neoplasias Esofágicas/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Terapia de Salvação , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Seguimentos , Humanos , Irinotecano/administração & dosagem , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Prognóstico , Quinazolinas/administração & dosagem , Estudos Retrospectivos , Taxa de Sobrevida , Tiofenos/administração & dosagem
7.
Virus Genes ; 55(5): 696-706, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31254238

RESUMO

Bacteriophages have been recently revisited as an alternative biocontrol tool due to the limitations of antibiotic treatment. In this study, we reported on the biological characteristics and genomic information of vB_KpnS_GH-K3 (abbreviated as GH-K3), a Klebsiella phage of the Siphoviridae family, which was previously isolated from a hospital sewage system. One-step growth curve analysis indicated that the burst size of GH-K3 was 291 PFU/cell. GH-K3 maintained a stable titer in a broad range of pH values (6-10) and temperature (up to 50 °C). Based on bioinformatics analysis, GH-K3 comprises of 49,427 bp containing a total of 77 open reading frames (ORFs), which share high degree of nucleotide similarity and close evolutionary relationships with at least 12 other Klebsiella phages. Of note, GH-K3 gp32 was identified as a unique ORF. The major segment of gp32 sequence at the C-terminus (residues 351-907) was found highly variable as determined by its mismatch with the nucleotide and protein sequences available at NCBI database. Furthermore, HHpred analysis indicated that GH-K3 gp32 contains three domains (PDB ID: 5W6S_A, 3GQ8_A and 1BHE_A) similar to depolymerase (depoKP36) of Klebsiella phage KP36 suggestive of a potential depolymerase activity during host receptor-binding in the processes of phage infection. Altogether, the current data revealed a novel putative depolymerase-like protein which is most likely to play an important role in phage-host interaction.


Assuntos
Bacteriófagos/crescimento & desenvolvimento , Klebsiella/virologia , Bacteriófagos/efeitos dos fármacos , Bacteriófagos/genética , Bacteriófagos/efeitos da radiação , Genoma Viral , Concentração de Íons de Hidrogênio , Viabilidade Microbiana/efeitos dos fármacos , Viabilidade Microbiana/efeitos da radiação , Fases de Leitura Aberta , Homologia de Sequência , Sintenia , Temperatura , Carga Viral , Proteínas Virais/genética
8.
Appl Environ Microbiol ; 84(21)2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30171001

RESUMO

Bacteriophage can be used as an alternative or complementary therapy to antibiotics for treating multidrug-resistant bacterial infections. However, the rapid emergence of resistant host variants during phage treatment has limited its therapeutic applications. In this study, a potential phage-resistant mechanism of Klebsiella pneumoniae was revealed. After phage GH-K3 treatment, a smooth-type colony, named K7RB, was obtained from the K. pneumoniae K7 culture. Treatment with IO4- and/or proteinase K indicated that polysaccharides of K7 played an important role in phage recruitment, and protein receptors on K7 were essential for effective infection by GH-K3. Differences in protein expression between K7 and K7RB were quantitatively analyzed by liquid chromatography-tandem mass spectrometry. Among differentially expressed proteins, OmpC, OmpN, KPN_02430, and OmpF were downregulated significantly in K7RBtrans-Complementation of OmpC in K7RB conferred rapid adsorption and sensitivity to GH-K3. In contrast, a single-base deletion mutation of ompC in K7, which resulted in OmpC silencing, led to lower adsorption efficiency and resistance to GH-K3. These assays proved that OmpC is the key receptor-binding protein for GH-K3. In addition, the native K. pneumoniae strains KPP14, KPP27, and KPP36 showed low or no sensitivity to GH-K3. However, these strains became more sensitive to GH-K3 after their native receptors were replaced by OmpC of K7, suggesting that OmpCK7 was the most suitable receptor for GH-K3. This study revealed that K7RB became resistant to GH-K3 due to gene mutation of ompC and that OmpC of K7 is essential for effective infection by GH-K3.IMPORTANCE With increased incidence of multidrug-resistant (MDR) bacterial strains, phages have regained attention as promising potential antibacterial agents. However, the rapid emergence of resistant variants during phage treatment has limited the therapeutic applications of phage. According to our trans-complementation, ompC mutation, and phage adsorption efficiency assays, we identified OmpC as the key receptor-binding protein (RBP) for phage GH-K3, which is essential for effective infection. This study revealed that the phage secondary receptor of K. pneumoniae, OmpC, is the essential RBP not only for phage infecting Gram-negative bacteria, such as Escherichia coli and Salmonella, but also for K. pneumoniae.


Assuntos
Bacteriófagos/fisiologia , Klebsiella pneumoniae/virologia , Porinas/metabolismo , Receptores Virais/metabolismo , Sequência de Aminoácidos , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Mutação , Porinas/genética , Receptores Virais/genética , Ligação Viral
9.
Appl Environ Microbiol ; 84(15)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29776929

RESUMO

Treatment of infections caused by staphylococci has become more difficult because of the emergence of multidrug-resistant strains as well as biofilm formation. In this study, we observed the ability of the phage lysin LysGH15 to eliminate staphylococcal planktonic cells and biofilms formed by Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus haemolyticus, and Staphylococcus hominis All these strains were sensitive to LysGH15, showing reductions in bacterial counts of approximately 4 log units within 30 min after treatment with 20 µg/ml of LysGH15, and the MICs ranged from 8 µg/ml to 32 µg/ml. LysGH15 efficiently prevented biofilm formation by the four staphylococcal species at a dose of 50 µg/ml. At a higher dose (100 µg/ml), LysGH15 also showed notable disrupting activity against 24-h and 72-h biofilms formed by S. aureus and coagulase-negative species. In the in vivo experiments, a single intraperitoneal injection of LysGH15 (20 µg/mouse) administered 1 h after the injection of S. epidermidis at double the minimum lethal dose was sufficient to protect the mice. The S. epidermidis cell counts were 4 log units lower in the blood and 3 log units lower in the organs of mice 24 h after treatment with LysGH15 than in the untreated control mice. LysGH15 reduced cytokine levels in the blood and improved pathological changes in the organs. The broad antistaphylococcal activity exerted by LysGH15 on planktonic cells and biofilms makes LysGH15 a valuable treatment option for biofilm-related or non-biofilm-related staphylococcal infections.IMPORTANCE Most staphylococcal species are major causes of health care- and community-associated infections. In particular, Staphylococcus aureus is a common and dangerous pathogen, and Staphylococcus epidermidis is a ubiquitous skin commensal and opportunistic pathogen. Treatment of infections caused by staphylococci has become more difficult because of the emergence of multidrug-resistant strains as well as biofilm formation. In this study, we found that all tested S. aureus, S. epidermidis, Staphylococcus haemolyticus, and Staphylococcus hominis strains were sensitive to the phage lysin LysGH15 (MICs ranging from 8 to 32 µg/ml). More importantly, LysGH15 not only prevented biofilm formation by these staphylococci but also disrupted 24-h and 72-h biofilms. Furthermore, the in vivo efficacy of LysGH15 was demonstrated in a mouse model of S. epidermidis bacteremia. Thus, LysGH15 exhibits therapeutic potential for treating biofilm-related or non-biofilm-related infections caused by diverse staphylococci.


Assuntos
Biofilmes , Terapia por Fagos , Plâncton/fisiologia , Plâncton/virologia , Infecções Estafilocócicas/terapia , Fagos de Staphylococcus/fisiologia , Staphylococcus/fisiologia , Staphylococcus/virologia , Animais , Bacteriemia/microbiologia , Bacteriemia/terapia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Plâncton/genética , Plâncton/crescimento & desenvolvimento , Infecções Estafilocócicas/microbiologia , Staphylococcus/genética , Staphylococcus/crescimento & desenvolvimento
10.
Virus Genes ; 54(3): 446-456, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29564689

RESUMO

Salmonella pullorum is the major pathogen that is harmful to the poultry industry in developing countries, and the treatment of chicken diarrhea caused by S. pullorum has become increasingly difficult. In this study, a virulent bacteriophage YSP2, which was able to specifically infect Salmonella, was isolated and characterized. Phage YSP2 was classified in the Siphoviridae family and had a short latent period of 10 min. No bacterial virulence- or lysogenesis-related ORF is present in the YSP2 genome, making it eligible for use in phage therapy. Experiments in vivo investigated the potential use of phages as a therapy against diarrhea in chickens caused by S. pullorum in a chicken diarrhea model, demonstrating that a single oral administration of YSP2 (1 × 1010 PFU/mL, 80 µL/chicken) 2 h after S. pullorum oral administration at a double median lethal dose was sufficient to protect chickens against diarrhea. Gross inspection showed that YSP2 can effectively reduce organ damage and significantly relieve hemorrhage in the intestine and liver tissue. Moreover, YSP2 can maintain a high curative effect when diluted to 108 PFU/mL. In light of its therapeutic effect on chicken diarrhea, YSP2 may serve as an alternative treatment strategy for infections caused by S. pullorum.


Assuntos
Galinhas , Diarreia/veterinária , Terapia por Fagos/veterinária , Doenças das Aves Domésticas/terapia , Salmonelose Animal/terapia , Fagos de Salmonella/isolamento & purificação , Animais , Diarreia/terapia , Feminino , Genoma Viral , Masculino , Salmonella , Fagos de Salmonella/fisiologia
11.
Appl Microbiol Biotechnol ; 102(2): 971-983, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29150707

RESUMO

In recent years, after the emergence of a large number of multidrug-resistant bacteria, phages and phage-associated products for the prevention and control of bacterial disease have revealed prominent advantages as compared with antibiotics. However, bacteria are susceptible to becoming phage-resistant, thus severely limiting the application of phage therapy. In this study, Escherichia coli cells were incubated with lytic bacteriophages to obtain mutants that were resistant to the lytic phages. Then, bacteriophages against the phage-resistant variants were isolated and subsequently mixed with the original lytic phage to prepare a novel phage cocktail for bactericidal use. The data showed that our phage cocktail not only had notable bactericidal effects, including a widened host range and rapid lysis, but also decreased the generation and mutation frequency of phage-resistant strains in vitro. In addition, we tested our cocktail in a murine bacteremia model. The results suggested that compared with the single phage, fewer phage-resistant bacteria appeared during the treatment of phage cocktail, thus prolonging the usable time of the phage cocktail and improving its therapeutic effect in phage applications. Importantly, our preparation method of phage cocktail was proved to be generalizable. Because the bacteriophage against the phage-resistant strain is an ideal guard that promptly attacks potential phage resistance, this guard-killer dual-function phage cocktail provides a novel strategy for phage therapy that allows the natural ecology to be sustained.


Assuntos
Bacteriólise , Bacteriófagos/fisiologia , Infecções por Escherichia coli/terapia , Escherichia coli/virologia , Terapia por Fagos , Animais , Escherichia coli/patogenicidade , Especificidade de Hospedeiro , Camundongos , Taxa de Mutação
12.
Antonie Van Leeuwenhoek ; 111(12): 2371-2384, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30008077

RESUMO

Actinobacillus pleuropneumoniae is the cause of porcine pleuropneumonia, for which the mortality rate is high. Host peripheral blood is a body site for the immune clearance of pathogens mediated by release of inflammatory factors. However, "out of control" inflammatory factor release can contribute to host death. To further understand the changes in the transcription level of immune-related effectors, samples of peripheral blood mononuclear cells (PBMCs) collected from piglets at different stages of infection (0, 24 and 120 h) were sequenced on an Illumina HiSeq™ 4000 platform. We found 3818 differentially expressed genes (DEGs) in the 24 h-infection group compared to the 0 h-infection group (Pb24-Vs-Pb0). DEGs mainly involved in the Gene ontology and KEGG pathways that included nucleic acid metabolism regulation, cell growth, cell differentiation, and organ morphological maintenance were not significantly enriched (P > 0.05). However, DEGs associated with protein kinase activity, receptor activation, metabolism, local adhesion and immune inflammatory responses were significantly enriched in Pb120-Vs-Pb24 (P < 0.05), as were those related to the T cell receptor signalling pathway, with most being down-regulated compared to the preceding stage (Pb24-Vs-Pb0). In PBMCs there were some changes in glucose metabolism, local adhesion and the immune inflammatory response (Pb120-Vs-Pb0). In addition, up-regulated DEGs, such as IL8, IL1ß, and CCL2, and were significantly enriched in immune-inflammatory related pathways compared to the uninfected stage, although they began to decline after 24 h.


Assuntos
Infecções por Actinobacillus/veterinária , Actinobacillus pleuropneumoniae/fisiologia , Leucócitos Mononucleares/imunologia , Pleuropneumonia/veterinária , Doenças dos Suínos/genética , Infecções por Actinobacillus/genética , Infecções por Actinobacillus/imunologia , Infecções por Actinobacillus/microbiologia , Animais , Feminino , Perfilação da Expressão Gênica , Leucócitos Mononucleares/microbiologia , Masculino , Pleuropneumonia/genética , Pleuropneumonia/imunologia , Pleuropneumonia/microbiologia , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/microbiologia
13.
J Gen Virol ; 97(5): 1272-1281, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26873847

RESUMO

Holins are phage-encoded hydrophobic membrane proteins that spontaneously and non-specifically accumulate and form lesions in the cytoplasmic membrane. The ORF72 gene (also designated HolGH15) derived from the genome of the Staphylococcus aureus phage GH15 was predicted to encode a membrane protein. An analysis indicated that the protein encoded by HolGH15 potentially consisted of two hydrophobic transmembrane helices. This protein exhibited the structural characteristics of class II holins and belonged to the phage_holin_1 superfamily. Expression of HolGH15 in Escherichia coli BL21 cells resulted in growth retardation of the host cells, which was triggered prematurely by the addition of 2,4-dinitrophenol. The expression of HolGH15 caused morphological alterations in engineered E. coli cells, including loss of the cell wall and cytoplasmic membrane integrity and release of intracellular components, which were visualized by transmission electron microscopy. HolGH15 exerted efficient antibacterial activity at 37 °C and pH 5.2. Mutation analysis indicated that the two transmembrane domains of HolGH15 were indispensable for the activity of the full-length protein. HolGH15 showed a broad antibacterial range: it not only inhibited Staphylococcus aureus, but also demonstrated antibacterial activity against other species, including Listeria monocytogenes, Bacillus subtilis, Pseudomonas aeruginosa, Klebsiella pneumoniae and E. coli. At the minimal inhibitory concentration, HolGH15 evoked the release of cellular contents and resulted in the shrinkage and death of Staphylococcus aureus and Listeria monocytogenes cells. To the best of our knowledge, this study is the first report of a Staphylococcus aureus phage holin that exerts antibacterial activity against heterogeneous pathogens.


Assuntos
Proteínas de Membrana/metabolismo , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/virologia , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Antibacterianos , Clonagem Molecular , Citoplasma , Regulação Viral da Expressão Gênica , Genoma Viral , Listeria monocytogenes , Proteínas de Membrana/genética , Mutação , Consumo de Álcool por Menores
14.
PLoS Pathog ; 10(5): e1004109, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24831957

RESUMO

The lysin LysGH15, which is derived from the staphylococcal phage GH15, demonstrates a wide lytic spectrum and strong lytic activity against methicillin-resistant Staphylococcus aureus (MRSA). Here, we find that the lytic activity of the full-length LysGH15 and its CHAP domain is dependent on calcium ions. To elucidate the molecular mechanism, the structures of three individual domains of LysGH15 were determined. Unexpectedly, the crystal structure of the LysGH15 CHAP domain reveals an "EF-hand-like" calcium-binding site near the Cys-His-Glu-Asn quartet active site groove. To date, the calcium-binding site in the LysGH15 CHAP domain is unique among homologous proteins, and it represents the first reported calcium-binding site in the CHAP family. More importantly, the calcium ion plays an important role as a switch that modulates the CHAP domain between the active and inactive states. Structure-guided mutagenesis of the amidase-2 domain reveals that both the zinc ion and E282 are required in catalysis and enable us to propose a catalytic mechanism. Nuclear magnetic resonance (NMR) spectroscopy and titration-guided mutagenesis identify residues (e.g., N404, Y406, G407, and T408) in the SH3b domain that are involved in the interactions with the substrate. To the best of our knowledge, our results constitute the first structural information on the biochemical features of a staphylococcal phage lysin and represent a pivotal step forward in understanding this type of lysin.


Assuntos
Cálcio/metabolismo , Mucoproteínas/química , Mucoproteínas/metabolismo , Domínios e Motivos de Interação entre Proteínas , Fagos de Staphylococcus/enzimologia , Amidoidrolases/química , Amidoidrolases/metabolismo , Sequência de Aminoácidos , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Domínio Catalítico , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Staphylococcus aureus Resistente à Meticilina/ultraestrutura , Testes de Sensibilidade Microbiana , Modelos Moleculares , Dados de Sequência Molecular , Mucoproteínas/farmacologia , Homologia de Sequência de Aminoácidos
15.
Appl Environ Microbiol ; 82(1): 87-94, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26475103

RESUMO

Pneumonia is one of the most prevalent Staphylococcus aureus-mediated diseases, and the treatment of this infection is becoming challenging due to the emergence of multidrug-resistant S. aureus, especially methicillin-resistant S. aureus (MRSA) strains. It has been reported that LysGH15, the lysin derived from phage GH15, displays high efficiency and a broad lytic spectrum against MRSA and that apigenin can markedly diminish the alpha-hemolysin of S. aureus. In this study, the combination therapy of LysGH15 and apigenin was evaluated in vitro and in a mouse S. aureus pneumonia model. No mutual adverse influence was detected between LysGH15 and apigenin in vitro. In animal experiments, the combination therapy showed a more effective treatment effect than LysGH15 or apigenin monotherapy (P < 0.05). The bacterial load in the lungs of mice administered the combination therapy was 1.5 log units within 24 h after challenge, whereas the loads in unprotected mice or mice treated with apigenin or LysGH15 alone were 10.2, 4.7, and 2.6 log units, respectively. The combination therapy group showed the best health status, the lowest ratio of wet tissue to dry tissue of the lungs, the smallest amount of total protein and cells in the lung, the fewest pathological manifestations, and the lowest cytokine level compared with the other groups (P < 0.05). With regard to its better protective efficacy, the combination therapy of LysGH15 and apigenin exhibits therapeutic potential for treating pneumonia caused by MRSA. This paper reports the combination therapy of lysin and natural products derived from traditional Chinese medicine.


Assuntos
Antibacterianos/administração & dosagem , Apigenina/administração & dosagem , Pneumonia/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Fagos de Staphylococcus/enzimologia , Staphylococcus aureus/efeitos dos fármacos , Proteínas Virais/administração & dosagem , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/microbiologia , Infecções Estafilocócicas/microbiologia , Fagos de Staphylococcus/química , Staphylococcus aureus/fisiologia
16.
BMC Microbiol ; 16(1): 171, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27473222

RESUMO

BACKGROUND: Brucella may establish chronic infection by regulating the expression of miRNAs. However, the role of miRNAs in modulating the intracellular growth of Brucella remains unclear. RESULTS: In this study, we show that Brucella. abortus infection leads to downregulation of miR-125b-5p in macrophages. We establish that miR-125b-5p targets A20, an inhibitor of the NF-kB activation. Additionally, expression of miR-125b-5p decreases A20 expression in B. abortus-infected macrophages and leads to NF-kB activation and increased production of TNFα. Furthermore, B. abortus survival is attenuated in the presence of miR-125b-5p. CONCLUSIONS: These results uncover a role for miR-125b-5p in the regulation of B. abortus intracellular survival via the control of A20 expression.


Assuntos
Brucella abortus/crescimento & desenvolvimento , Brucella abortus/genética , Brucelose/microbiologia , Citoplasma/efeitos dos fármacos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Sobrevida/fisiologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/efeitos dos fármacos , Animais , Brucella abortus/metabolismo , Citoplasma/metabolismo , Regulação para Baixo , Regulação Bacteriana da Expressão Gênica , Ativação de Macrófagos , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Células RAW 264.7/microbiologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
17.
Arch Microbiol ; 198(9): 923-31, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27286866

RESUMO

The intracellular pathogen Brucella abortus (B. abortus) survives and replicates inside host cells within the Brucella-containing vacuole, in which membrane contains a small GTPase Rab1. Here, we reported that Rab1 mediates B. abortus intracellular growth. Furthermore, B. abortus DnaK was identified to interact with Rab1 using GST pull-down and mass spectrometry analysis. This interaction was confirmed by co-immunoprecipitation and immunofluorescence. Through DnaK-CyaA fusion protein translocation assay and immunofluorescence confocal microscopy, the B. abortus DnaK was proved to be a virB-dependent translocated substrate.


Assuntos
Proteínas de Bactérias/fisiologia , Brucella abortus/fisiologia , Sobrevivência Celular/fisiologia , Proteínas rab1 de Ligação ao GTP/fisiologia , Proteínas de Bactérias/metabolismo , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/patogenicidade , Interações Hospedeiro-Patógeno/fisiologia , Ligação Proteica/fisiologia , Transporte Proteico/fisiologia , Vacúolos/microbiologia
18.
Antonie Van Leeuwenhoek ; 109(1): 51-70, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26494209

RESUMO

Actinobacillus pleuropneumoniae is an important pathogen that causes respiratory disease in pigs. Trimeric autotransporter adhesin (TAA) is a recently discovered bacterial virulence factor that mediates bacterial adhesion and colonization. Two TAA coding genes have been found in the genome of A. pleuropneumoniae strain 5b L20, but whether they contribute to bacterial pathogenicity is unclear. In this study, we used homologous recombination to construct a double-gene deletion mutant, ΔTAA, in which both TAA coding genes were deleted and used it in in vivo and in vitro studies to confirm that TAAs participate in bacterial auto-aggregation, biofilm formation, cell adhesion and virulence in mice. A microarray analysis was used to determine whether TAAs can regulate other A. pleuropneumoniae genes during interactions with porcine primary alveolar macrophages. The results showed that deletion of both TAA coding genes up-regulated 36 genes, including ene1514, hofB and tbpB2, and simultaneously down-regulated 36 genes, including lgt, murF and ftsY. These data illustrate that TAAs help to maintain full bacterial virulence both directly, through their bioactivity, and indirectly by regulating the bacterial type II and IV secretion systems and regulating the synthesis or secretion of virulence factors. This study not only enhances our understanding of the role of TAAs but also has significance for those studying A. pleuropneumoniae pathogenesis.


Assuntos
Actinobacillus pleuropneumoniae/patogenicidade , Adesinas Bacterianas/genética , Regulação Bacteriana da Expressão Gênica , Macrófagos Alveolares/microbiologia , Infecções por Actinobacillus/microbiologia , Actinobacillus pleuropneumoniae/genética , Actinobacillus pleuropneumoniae/metabolismo , Actinobacillus pleuropneumoniae/fisiologia , Adesinas Bacterianas/metabolismo , Animais , Aderência Bacteriana/genética , Aderência Bacteriana/fisiologia , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Genes Bacterianos , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Cultura Primária de Células , Suínos , Sistemas de Secreção Tipo V/genética , Sistemas de Secreção Tipo V/metabolismo , Fatores de Virulência/biossíntese , Fatores de Virulência/genética
19.
Int J Clin Pract ; 70 Suppl 9B: B56-63, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27577516

RESUMO

OBJECTIVE: The aim of this study was to evaluate the radiation dose and renal cell carcinoma conspicuity with virtual unenhanced images and split-bolus injection from spectral multidetector CT (MDCT). MATERIAL AND METHODS: This prospective study was approved by the Ethics Committee, and informed consent was obtained. Ninety suspected patients of renal cell carcinoma diagnosed by abdominal ultrasonography and CEUS were randomly divided into two groups by a radiographer. Patients of the first group underwent spectral MDCT with virtual unenhanced imaging and split-bolus injection, while patients in the second group underwent conventional unenhanced as well as tri-phasic enhanced CT. Group A (split-bolus spectral MDCT group): The contrast material was administered at a dose of 1.5 mL/kg body weight at a flow rate of 4 mL/s, with a ratio of 7 to 5 before the CT scan with an interval of 60 seconds. Virtual unenhanced images were generated using a standard three-material decomposition algorithm, and the best mono-energy (keV) was calculated to show the tumour, renal artery and renal vein. Group B (conventional tri-phasic enhanced CT group): the contrast agent was injected with a dose of 1.5 mL/kg body weight at a flow rate of 4 mL/s. The corticomedullary phase scanning was performed once the arterial CT value reached 100 HU; the nephrographic phase was scanned 60 seconds later. And the excretory phase was scanned 5 min after onset of contrast injection. The LKR (lesion kidney ratio), CNR, and CT value of the corticomedullary and nephrographic phase were measured. The opacification of the renal collection system (including calices, infundibula and renal pelvis) was scored. The radiation dose was recorded. Statistical analysis was performed using Student's t-test, Fisher's exact test, the Mann-Whitney U-test, and k statistics. RESULTS: There were no statistically significant differences between the two groups in age, sex and body mass index (BMI), but there was significant difference in treatment methods. The best mono-energy was 58 keV for showing the tumour and renal artery and 67 keV for showing the renal vein. There were no differences in the mean attenuations of normal renal parenchyma, renal tumour, CNR, and imaging quality between true unenhanced images and virtual unenhanced images from the combined corticomedullary and nephrographic phase. The LKR of the mono-energy at 58 keV from the combined corticomedullary and nephrographic phase was significantly better than the corticomedullary phase of the conventional enhanced CT scan (0.74±0.18 vs 1.08±0.34, P<.01), but there was no difference in CNR (2.31±1.74 vs 2.79±1.83, P>.05). There were no differences in the CT values of the renal tumour, normal renal parenchyma and renal artery between the two groups (P>.05). The CT value of the renal vein at mono-energy (67 keV) (200.55±43.38) from the combined corticomedullary and nephrographic phase was higher than the conventional CT scan (140.90±42.64) in the nephrographic phase. The Kappa scores of the rate of the renal collection system for the conventional CT and spectral CT were 0.68 (95% confidence interval [CI]: 0.35-0.89) and 0.54 (95% CI: 0.30-0.88), respectively. The radiation dose (735±162 mGy·cm) of Group A was significantly less than that of Group B (1032±324 mGy·cm) (P<.01). CONCLUSION: Conspicuity with virtual unenhanced imaging and split-bolus injection from spectral multidetector CT is better than or equal to the conventional three-phase enhanced CT scan in showing the RCC, renal artery and renal vein, while the radiation dose can be reduced by 28.78%.


Assuntos
Carcinoma de Células Renais/diagnóstico por imagem , Meios de Contraste/administração & dosagem , Neoplasias Renais/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Adulto , Idoso , Feminino , Humanos , Rim/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Doses de Radiação , Artéria Renal/diagnóstico por imagem , Veias Renais/diagnóstico por imagem
20.
Microb Pathog ; 89: 177-83, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26523973

RESUMO

Brucella DNA can be sensed by TLR9 on endosomal membrane and by cytosolic AIM2-inflammasome to induce proinflammatory cytokine production that contributes to partially activate innate immunity. Additionally, Brucella DNA has been identified to be able to act as a major bacterial component to induce type I IFN. However, the role of Brucella DNA in Brucella intracellular growth remains unknown. Here, we showed that stimulation with Brucella DNA promote macrophage activation in TLR9-dependent manner. Activated macrophages can suppresses wild type Brucella intracellular replication at early stage of infection via enhancing NO production. We also reported that activated macrophage promotes bactericidal function of macrophages infected with VirB-deficient Brucella at the early or late stage of infection. This study uncovers a novel function of Brucella DNA, which can help us further elucidate the mechanism of Brucella intracellular survival.


Assuntos
Brucella/crescimento & desenvolvimento , Brucella/imunologia , DNA Bacteriano/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/microbiologia , Óxido Nítrico/metabolismo , Animais , Antibacterianos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/deficiência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa