Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Virulence ; 15(1): 2306719, 2024 12.
Artigo em Inglês | MEDLINE | ID: mdl-38251714

RESUMO

The arginine deiminase system (ADS) has been identified in various bacteria and functions to supplement energy production and enhance biological adaptability. The current understanding of the regulatory mechanism of ADS and its effect on bacterial pathogenesis is still limited. Here, we found that the XRE family transcriptional regulator XtrSs negatively affected Streptococcus suis virulence and significantly repressed ADS transcription when the bacteria were incubated in blood. Electrophoretic mobility shift (EMSA) and lacZ fusion assays further showed that XtrSs directly bind to the promoter of ArgR, an acknowledged positive regulator of bacterial ADS, to repress ArgR transcription. Moreover, we provided compelling evidence that S. suis could utilize arginine via ADS to adapt to acid stress, while ΔxtrSs enhanced this acid resistance by upregulating the ADS operon. Moreover, whole ADS-knockout S. suis increased arginine and antimicrobial NO in the infected macrophage cells, decreased intracellular survival, and even caused significant attenuation of bacterial virulence in a mouse infection model, while ΔxtrSs consistently presented the opposite results. Our experiments identified a novel ADS regulatory mechanism in S. suis, whereby XtrSs regulated ADS to modulate NO content in macrophages, promoting S. suis intracellular survival. Meanwhile, our findings provide a new perspective on how Streptococci evade the host's innate immune system.


Assuntos
Proteínas de Bactérias , Infecções Estreptocócicas , Streptococcus suis , Animais , Camundongos , Arginina , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Hidrolases/genética , Hidrolases/metabolismo , Macrófagos , Infecções Estreptocócicas/microbiologia , Streptococcus suis/patogenicidade , Streptococcus suis/fisiologia
2.
Vet Microbiol ; 251: 108925, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33181436

RESUMO

Streptococcus suis (S. suis) is an emerging zoonotic pathogen that can cause meningitis, arthritis, pneumonia, and sepsis. It poses a serious threat to the swine industry and public health worldwide. Ornithine carbamoyltransferase (OTC) is involved in the arginine deiminase system. OTC, which is a widely distributed enzyme in microorganisms, mammals, and higher plants, catalyzes the conversion of ornithine to citrulline. The present study showed that the otc gene plays an important role in the pathogenesis of S. suis infections. The ability of an otc-deficient mutant (Δotc) to form a biofilm was significantly reduced compared to the wild-type (WT) strain, as determined by crystal violet staining. Confocal laser scanning microscopy and scanning electron microscopy observations showed that the weakening of biofilm formation by the Δotc strain is related to a decrease in the extracellular matrix. In addition, compared to the WT strain, the Δotc strain had a reduced capacity to adhere to human laryngeal epidermoid carcinoma (HEp-2) cells compared to the WT strain. A real-time PCR analysis showed that the expression of adhesion-related genes by the Δotc strain was also lower than that of the WT strain. The virulence of the Δotc strain was significantly lower than that of the WT strain in a murine infection model. In addition, a histological analysis showed that the pathogenicity of the Δotc strain was lower than that of the WT strain, causing only slight inflammatory lesions in lung, liver, spleen, and kidney tissues. No significant differences were observed between the complemented mutant (CΔotc) and WT strains with respect to biofilm formation, adhesion, gene expression, and virulence. The present study provided evidence that the otc gene plays a pivotal role in the regulation of S. suis adhesion and biofilm formation. It also suggested that the otc gene is indirectly involved in the pathogenesis of S. suis serotype 2 infections.


Assuntos
Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Biofilmes/crescimento & desenvolvimento , Ornitina Carbamoiltransferase/genética , Infecções Estreptocócicas/veterinária , Streptococcus suis/genética , Streptococcus suis/patogenicidade , Fatores de Virulência/genética , Animais , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Organismos Livres de Patógenos Específicos , Infecções Estreptocócicas/virologia , Streptococcus suis/fisiologia , Suínos , Virulência
3.
Vet Res ; 51(1): 31, 2020 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-32106883

RESUMO

Porcine circovirus type 2 (PCV2) is considered as the primary pathogen of porcine circovirus-associated disease (PCVAD), which results in significant economic losses worldwide. Clinically, PCV2 often causes disease through coinfection with other bacterial pathogens, including Streptococcus suis (S. suis), and especially the highly prevalent S. suis serotype 2 (SS2). The present study determined that continuous PCV2 infection in piglets down-regulates tight junction proteins (TJ) ZO-1 and occludin in the lungs. Swine tracheal epithelial cells (STEC) were used to explore the mechanisms and consequences of disruption of TJ, and an in vitro tracheal epithelial barrier model was established. Our results show that PCV2 infection in STEC decreases the expression levels of ZO-1 and occludin and increases the permeability of the tracheal epithelial barrier, resulting in easier translocation of SS2. Moreover, Western blot analysis indicates that PCV2 infection activates the JNK/MAPK pathway. The disruption of TJ in SETC and increased permeability of the epithelial barrier induced by PCV2 could be alleviated by inhibition of JNK phosphorylation, which indicates that the JNK/MAPK pathway regulates the expression of ZO-1 and occludin during PCV2 infection. This study allows us to better understand the mechanisms of PCV2 coinfection with bacterial pathogens and provides new insight into controlling the occurrence of PCVAD.


Assuntos
Infecções por Circoviridae/veterinária , Circovirus/fisiologia , Coinfecção/veterinária , Transdução de Sinais , Infecções Estreptocócicas/veterinária , Streptococcus suis/fisiologia , Doenças dos Suínos/microbiologia , Animais , Linhagem Celular , Infecções por Circoviridae/virologia , Coinfecção/microbiologia , Coinfecção/virologia , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Infecções Estreptocócicas/microbiologia , Suínos , Doenças dos Suínos/virologia , Junções Íntimas , Traqueia/microbiologia , Traqueia/virologia
4.
Appl Environ Microbiol ; 85(9)2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30824438

RESUMO

Nursing mother and breed can differently regulate early-life microbiota succession in pigs. However, it remains unclear whether they affect gastrointestinal microbiota and immune status, which are critical for early-life gut health. Here, an interspecific cross-fostering piglet model was employed by fostering neonatal Yorkshire and Meishan piglets to the same or another breed of sows. Jejunal and colonic microbiotas and mucosal immune parameters were analyzed at postnatal days 14 (preweaning) and 49 (postweaning). Nursing mother affected 10 genera in the colon and 3 minor genera in the jejunum. At day 14, Meishan sow-nursed piglets had lower Streptococcus suis and higher Cloacibacillus counts in the colonic digesta and larger amounts of interleukin 10 and Foxp3-positive cells in the colonic mucosa than did Yorkshire sow-nursed piglets. At day 49, nursing mother had no significant effects on cytokine expression. Breed effects were observed; Meishan piglets had lower relative abundances of Prevotella and lower gene expression of tumor necrosis factor alpha (TNF-α) than those of Yorkshire piglets at days 14 and 49. Collectively, nursing mother mainly affected preweaning colonic microbiota and immune status, while breed effects persisted after weaning. Piglets nursed by Meishan sows had different microbiota compositions and inflammatory cytokine profiles in the colon compared with those of piglets nursed by Yorkshire sows. These results highlight the different role of nursing mother and breed in affecting early gut microenvironment.IMPORTANCE Early-life gut microbiota and immune status are pivotal for postnatal growth. By using an interspecific cross-fostering piglet model, we find that change in nursing mother transiently reshapes preweaning colon microbiota and immune status, while breed shows persistent effects both pre- and postweaning. Piglets nursed by Meishan sows had lower Streptococcus suis counts and higher anti-inflammatory cytokine expression. These results highlight the significance of nursing mother in regulating early-life gut health.


Assuntos
Colo/microbiologia , Microbioma Gastrointestinal/fisiologia , Imunidade Inata/fisiologia , Sus scrofa/fisiologia , Animais , Citocinas/metabolismo , Feminino , Especificidade da Espécie , Streptococcus suis/fisiologia , Sus scrofa/genética , Sus scrofa/imunologia , Sus scrofa/microbiologia , Desmame
5.
Artigo em Inglês | MEDLINE | ID: mdl-30863725

RESUMO

Streptococcosis is recognized as a leading infectious disease in the swine industry. Streptococcus suis serotype 2 is regarded as the most virulent species, which threatens human and pig health and causes serious economic losses. In this study, multiple in vitro and in vivo effects of MP1102 on multidrug resistant S. suis was studied for the first time. MP1102 exhibited significant antibacterial activity against S. suis (minimum inhibitory concentration, MIC = 0.028-0.228 µM), rapid bacteriocidal action, a longer postantibiotic effect than ceftriaxone, and a synergistic or additive effect with lincomycin, penicillin, and ceftriaxone (FICI = 0.29-0.96). No resistant mutants appeared after 30 serial passages of S. suis in the presence of MP1102. Flow cytometric analysis and electron microscopy observations showed that MP1102 destroyed S. suis cell membrane integrity and affected S. suis cell ultrastructure and membrane morphology. Specifically, a significantly wrinkled surface, intracellular content leakage, and cell lysis were noted, establishing a cyto-basis of nonresistance to this pathogen. DNA gel retardation and circular dichroism analysis indicated that MP1102 interacted with DNA by binding to DNA and changing the DNA conformation, even leading to the disappearance of the helical structure. This result further supported the mechanistic basis of nonresistance via interaction with an intracellular target, which could serve as a means of secondary injury after MP1102 is transported across the membrane. Upon treatment with 2.5-5.0 mg/kg MP1102, the survival of mice challenged with S. suis was 83.3-100%. MP1102 decreased bacterial translocation in liver, lung, spleen, and blood; inhibited the release of interleukin-1ß and tumor necrosis factor-α; and relieved the lung, liver, and spleen from acute injury induced by S. suis. These results suggest that MP1102 is a potent novel antibacterial agent for the treatment of porcine streptococcal disease.


Assuntos
Antibacterianos/administração & dosagem , Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla , Infecções Estreptocócicas/tratamento farmacológico , Infecções Estreptocócicas/microbiologia , Streptococcus suis/efeitos dos fármacos , Estruturas Animais/microbiologia , Estruturas Animais/patologia , Animais , Bacteriólise/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/ultraestrutura , DNA Bacteriano/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Camundongos , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Conformação de Ácido Nucleico/efeitos dos fármacos , Sorogrupo , Infecções Estreptocócicas/patologia , Streptococcus suis/classificação , Streptococcus suis/fisiologia , Streptococcus suis/ultraestrutura , Análise de Sobrevida
6.
Int Immunopharmacol ; 69: 71-78, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30682719

RESUMO

Streptococcus suis, a globally distributed bacterial pathogen, is an important zoonotic agent for humans and animals that can lead to multiple deaths and cause major economic losses. Suilysin (SLY), secreted by most pathogenic S. suis strains, is a cytotoxic toxin that belongs to the cholesterol-dependent cytolysin family; this toxin plays a key role in a mouse meningitis model, suggesting that effective interference with the biological activity of SLY may be a potential treatment for S. suis infection. In addition, the inflammatory response induced by S. suis is an important manifestation in infections and is associated with multiple fatal diseases. In this study, we found that the natural compound quercetin can directly inhibit the pore-forming activity of SLY without affecting bacterial growth and SLY secretion at the concentrations tested in our assay. In addition, quercetin treatment significantly alleviated cytotoxicity caused by S. suis infection and effectively reduced the release of the pro-inflammatory cytokines IL-1ß, IL-6, and tumor necrosis factor alpha (TNF-α) stimulated by bacteria. Significantly decreased mortality was observed for the S. suis-infected mice that received quercetin. Our results suggested that quercetin may represent a promising therapeutic candidate for S. suis infection by targeting SLY and the subsequent inflammation. The present study provides a new strategy and leading compound for S. suis infection.


Assuntos
Anti-Inflamatórios/uso terapêutico , Proteínas Hemolisinas/metabolismo , Macrófagos/efeitos dos fármacos , Quercetina/uso terapêutico , Infecções Estreptocócicas/tratamento farmacológico , Streptococcus suis/fisiologia , Doenças dos Suínos/tratamento farmacológico , Animais , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Streptococcus suis/patogenicidade , Suínos , Virulência
7.
Microb Pathog ; 122: 90-97, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29886087

RESUMO

Streptococcus suis serotype 2 (SS2) is an important zoonotic pathogen that infects swine and humans with high mortality and morbidity. Although a number of virulence-associated factors have been reported, the understanding of the molecular mechanism underlying SS2 pathogenicity remains limited. Our previous studies revealed that srtBCD-associated protein 2' (SBP2') contributed to the pathogenesis of SS2, but the function of another member in the srtBCD cluster, srtBCD-associated protein 1 (SBP1) was still unknown. Here, we found that sbp1 was widely distributed among high virulent SS2 strains, suggesting that sbp1 may be involved in the pathogenesis of SS2. To investigate the function of SBP1, we firstly conducted Western blotting analyses to confirm that SBP1 was expressed in the high virulent SS2 strain ZY05719 both in vivo and in vitro, then constructed the deletion mutant of sbp1 by homologous recombination. Bacterial adhesion assay, indirect immunofluorescence assay and protein binding assay all demonstrated that SBP1 was associated with adhesion of SS2 to HEp-2 cells. However, SBP1 did not influence the invasion, phagocytosis or intracellular survival of SS2. Furthermore, infection assays in vivo showed that inactivation of sbp1 failed to impair the ability of SS2 to cause zebrafish and mouse mortality. Overall, these results indicate that SBP1 is an adhesion-associated factor without the involvement of virulence in Streptococcus suis serotype 2.


Assuntos
Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Streptococcus suis/fisiologia , Adesinas Bacterianas/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Modelos Animais de Doenças , Endocitose , Células Epiteliais/microbiologia , Deleção de Genes , Perfilação da Expressão Gênica , Humanos , Camundongos , Viabilidade Microbiana , Fagocitose , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Virulência , Peixe-Zebra
8.
Ann Agric Environ Med ; 25(1): 186-203, 2018 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-29575852

RESUMO

Streptococcus suis is a re-emerging zoonotic pathogen that may cause severe disease, mostly meningitis, in pigs and in humans having occupational contact with pigs and pork, such as farmers, slaughterhose workers and butchers. The first stage of the pathogenic process, similar in pigs and humans, is adherence to and colonisation of mucosal and/or epithelial surface(s) of the host. The second stage is invasion into deeper tissue and extracellular translocation of bacterium in the bloodstream, either free in circulation or attached to the surface of monocytes. If S. suis present in blood fails to cause fatal septicaemia, it is able to progress into the third stage comprising penetration into host's organs, mostly by crossing the blood-brain barrier and/or blood-cerebrospinal fluid barrier to gain access to the central nervous system (CNS) and cause meningitis. The fourth stage is inflammation that plays a key role in the pathogen esis of both systemic and CNS infections caused by S. suis. The pathogen may induce the overproduction of pro-inflammatory cytokines that cause septic shock and/or the recruitment and activation of different leukocyte populations, causing acute inflammation of the CNS. Streptococcus suis can also evoke - through activation of microglial cells, astrocytes and possibly other cell types - a fulminant inflammatory reaction of the brain which leads to intracranial complications, including brain oedema, increased intracranial pressure, cerebrovascular insults, and deafness, as a result of cochlear sepsis. In all stages of the pathogenic process, S. suis interacts with many types of immunocompetent host's cells, such as polymorphonuclear leukocytes, mononuclear macrophages, lymphocytes, dendritic cells and microglia, using a range of versatile virulence factors for evasion of the innate and adaptive immune defence of the host, and for overcoming environmental stress. It is estimated that S. suis produces more than 100 different virulence factors that could be classified into 4 groups: surface components or secreted elements, enzymes, transcription factors or regulatory systems and transporter factors or secretion systems. A major virulence factor is capsular polysaccharide (CPS) that protects bacteria from phagocytosis. However, it hampers adhesion to and invasion of host's cells, release of inflammatory cytokines and formation of the resistant biofilm which, in many cases, is vital for the persistence of bacteria. It has been demonstrated that the arising by mutation unencapsulated S. suis clones, which are more successful in penetration to and propagation within the host's cells, may coexist in the organism of a single host together with those that are encapsulated. Both 'complementary' clones assist each other in the successful colonization of host's tissues and persistence therein. S. suis has an open pan-genome characterized by a frequent gene transfer and a large diversity. Of the genetic determinants of S. suis pathogenicity, the most important are pathogenicity islands (PAI), in particular, a novel DNA segment of 89 kb length with evident pathogenic traits that has been designated as 89K PAI. It has been estimated that more than one-third of the S. suis virulence factors is associated with this PAI. It has been proved that the virulent S. suis strains possess smaller genomes, compared to avirulent ones, but more genes associated with virulence. Overall, the evolution of the species most probably aims towards increased pathogenicity, and hence the most significant task of the current research is an elaboration of a vaccine, efficient both for humans and pigs.


Assuntos
Doenças dos Trabalhadores Agrícolas/microbiologia , Exposição Ocupacional/efeitos adversos , Infecções Estreptocócicas/microbiologia , Streptococcus suis/fisiologia , Doenças dos Suínos/microbiologia , Zoonoses/microbiologia , Animais , Humanos , Infecções Estreptocócicas/transmissão , Streptococcus suis/genética , Streptococcus suis/isolamento & purificação , Streptococcus suis/patogenicidade , Suínos , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Zoonoses/transmissão
9.
Artigo em Inglês | MEDLINE | ID: mdl-29479521

RESUMO

Streptococcus suis serotype 2 (SS2) is a zoonotic pathogen that can cause meningitis both in pigs and in human beings. However, the pathogenesis of central nervous system (CNS) infection caused by SS2 have not yet been elucidated. To find the key molecules in cerebrospinal fluid (CSF) needed for the pathogenesis, a SS2 meningoencephalitic pig model and a SS2 non-meningoencephalitic pig model were established in this study. CSF was collected from infected piglets, and protein profiling was performed with label-free proteomics technology. A total of 813 differential proteins, including 52 up-regulated proteins and 761 down-regulated proteins, were found in the CSF of meningoencephalitic pigs compared with both non-meningoencephalitic pigs and healthy pigs. These 813 differential proteins were clustered into three main categories, namely, cellular component, biological process, and molecular function by gene ontology (GO) analysis. The most enriched subclasses of differential proteins in each category were exosome (44.3%), energy pathway (25.0%) and catalytic activity (11.3%), respectively. The most enriched subclasses of upregulated proteins were extracellular (62.1%), protein metabolism (34.5%) and cysteine-type peptidase activity (6.9%), and of downregulated proteins were exosomes (45.0%), energy pathway (24.0%) and catalytic activity (9.4%). Then, the differential proteins were further investigated by using the KEGG database and were found to participate in 16 KEGGs. The most enriched KEGG was citrate cycle (56.6%), and some of these differential proteins are associated with brain diseases such as Huntington's disease (18.6%), Parkinson's disease (23.8%) and Alzheimer's disease (17.6%). Sixteen of the 813 differential proteins, chosen randomly as examples, were further confirmed by enzyme-linked immunosorbent assay (ELISA) to support the proteomic data. To our knowledge, this is the first study to analyze the differential protein profiling of CSF between SS2 meningoencephalitic piglets and non-meningoencephalitic piglets by employing proteomic technology. The discovery and bioinformatics analysis of these differential proteins provides reference data not only for research on pathogenesis of SS2 CNS infection but also for diagnosis and drug therapy research.


Assuntos
Meningoencefalite/veterinária , Proteoma , Proteômica , Infecções Estreptocócicas/veterinária , Streptococcus suis/fisiologia , Doenças dos Suínos/líquido cefalorraquidiano , Doenças dos Suínos/microbiologia , Animais , Estudos de Casos e Controles , Cromatografia Líquida , Biologia Computacional/métodos , Ontologia Genética , Anotação de Sequência Molecular , Proteômica/métodos , Streptococcus suis/classificação , Suínos , Doenças dos Suínos/diagnóstico , Espectrometria de Massas em Tandem
10.
Artigo em Inglês | MEDLINE | ID: mdl-30687645

RESUMO

Bacterial penetration of the blood-brain barrier requires its successful invasion of brain microvascular endothelial cells (BMECs), and host actin cytoskeleton rearrangement in these cells is a key prerequisite for this process. We have reported previously that meningitic Escherichia coli can induce the activation of host's epidermal growth factor receptor (EGFR) to facilitate its invasion of BMECs. However, it is unknown how EGFR specifically functions during this invasion process. Here, we identified an important EGFR-interacting protein, α-actinin-4 (ACTN4), which is involved in maintaining and regulating the actin cytoskeleton. We observed that transactivated-EGFR competitively recruited ACTN4 from intracellular F-actin fibers to disrupt the cytoskeleton, thus facilitating bacterial invasion of BMECs. Strikingly, this mechanism operated not only for meningitic E. coli, but also for infections with Streptococcus suis, a Gram-positive meningitis-causing bacterial pathogen, thus revealing a common mechanism hijacked by these meningitic pathogens where EGFR competitively recruits ACTN4. Ever rising levels of antibiotic-resistant bacteria and the emergence of their extended-spectrum antimicrobial-resistant counterparts remind us that EGFR could act as an alternative non-antibiotic target to better prevent and control bacterial meningitis.


Assuntos
Actinina/metabolismo , Endocitose , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Receptores ErbB/metabolismo , Escherichia coli/fisiologia , Streptococcus suis/fisiologia , Células Cultivadas , Humanos
11.
Ann Agric Environ Med ; 24(4): 683-695, 2017 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-29284248

RESUMO

Streptococcus suis (ex Elliot 1966, Kilpper-Bälz & Schleifer 1987) is a facultatively anaerobic Gram-positive ovoid or coccal bacterium surrounded by a polysaccharide capsule. Based on the antigenic diversity of the capsule, S. suis strains are classified serologically into 35 serotypes. Streptococcus suis is a commensal of pigs, commonly colonizing their tonsils and nasal cavities, mostly in weaning piglets between 4-10 weeks of age. This species occurs also in cattle and other mammals, in birds and in humans. Some strains, mostly those belonging to serotype 2, are also pathogenic for pigs, as well as for other animals and humans. Meningitis is the primary disease syndrome caused by S. suis, both in pigs and in humans. It is estimated that meningitis accounted for 68.0% of all cases of human disease reported until the end of 2012, followed by septicaemia (including life-threatening condition described as 'streptococcal toxic shock-like syndrome' - STSLS), arthritis, endocarditis, and endophthalmitis. Hearing loss and/or ves tibular dysfunction are the most common sequelae after recovery from meningitis caused by S. suis, occurring in more than 50% of patients. In the last two decades, the number of reported human cases due to S. suis has dramatically increased, mostly due to epidemics recorded in China in 1998 and 2005, and the fulminant increase in morbidity in the countries of south-eastern Asia, mostly Vietnam and Thailand. Out of 1,642 cases of S. suis infections identified between 2002-2013 worldwide in humans, 90.2% occurred in Asia, 8.5% in Europe and 1.3% in other parts of the globe. The human disease has mostly a zoonotic and occupational origin and occurs in pig breeders, abattoir workers, butchers and workers of meat processing facilities, veterinarians and meat inspectors. Bacteria are transmitted to workers by close contact with pigs or pig products, usually through contamination of minor cuts or abrasions on skin of hands and/or arms, or by pig bite. A different epidemiologic situation occurs in the Southeast Asian countries where most people become infected by habitual consumption of raw or undercooked pork, blood and offal products in the form of traditional dishes. Prevention of S. suis infections in pigs includes vaccination, improvement in pig-raising conditions, disinfection and/or fumigation of animal houses, and isolation of sick animals at the outbreak of disease. Prevention of human infections comprises: protection of skin from pig bite or injury with sharp tools by people occupationally exposed to pigs and pig products, prompt disinfection and dressing of wounds and abrasions at work, protection of the respiratory tract by wearing appropriate masks or repirators, consulting a doctor in the case of febrile illness after exposure to pigs or pork meat, avoidance of occupations associated with exposure to pigs and pork by immunocompomised people, avoidance of consumption of raw pork or pig blood, adequate cooking of pork, and health education.


Assuntos
Produtos da Carne/microbiologia , Exposição Ocupacional/análise , Infecções Estreptocócicas/epidemiologia , Infecções Estreptocócicas/veterinária , Streptococcus suis/fisiologia , Doenças dos Suínos/epidemiologia , Matadouros , Animais , China , Humanos , Produtos da Carne/análise , Infecções Estreptocócicas/microbiologia , Streptococcus suis/genética , Streptococcus suis/isolamento & purificação , Suínos , Doenças dos Suínos/microbiologia , Zoonoses/epidemiologia , Zoonoses/microbiologia
12.
Artigo em Inglês | MEDLINE | ID: mdl-28373968

RESUMO

Invasive infections caused by Streptococcus suis serotype 2 (SS2) has emerged as a clinical problem in recent years. Neutrophil extracellular traps (NETs) are an important mechanism for the trapping and killing of pathogens that are resistant to phagocytosis. Biofilm formation can protect bacteria from being killed by phagocytes. Until now, there have only been a few studies that focused on the interactions between bacterial biofilms and NETs. SS2 in both a biofilm state and a planktonic cell state were incubated with phagocytes and NETs, and bacterial survival was assessed. DNase I and cytochalasin B were used to degrade NET DNA or suppress phagocytosis, respectively. Extracellular DNA was stained with impermeable fluorescent dye to quantify NET formation. Biofilm formation increased up to 6-fold in the presence of neutrophils, and biofilms were identified in murine tissue. Both planktonic and biofilm cells induced neutrophils chemotaxis to the infection site, with neutrophils increasing by 85.1 and 73.8%, respectively. The bacteria in biofilms were not phagocytized. The bactericidal efficacy of NETs on the biofilms and planktonic cells were equal; however, the biofilm extracellular matrix can inhibit NET release. Although biofilms inhibit NETs release, NETs appear to be an important mechanism to eliminate SS2 biofilms. This knowledge advances the understanding of biofilms and may aid in the development of treatments for persistent infections with a biofilm component.


Assuntos
Biofilmes/crescimento & desenvolvimento , Armadilhas Extracelulares/metabolismo , Interações Hospedeiro-Patógeno , Neutrófilos/imunologia , Sorogrupo , Streptococcus suis/imunologia , Streptococcus suis/fisiologia , Animais , Células Cultivadas , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Viabilidade Microbiana , Neutrófilos/microbiologia , Fagocitose
13.
J Neuroinflammation ; 13(1): 274, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27756321

RESUMO

BACKGROUND: Streptococcus suis serotype 2 (SS2) is an important zoonotic bacterial pathogen in both humans and animals, which can cause high morbidity and mortality. Meningitis is one of the major clinical manifestations of SS2 infection. However, the specific process of SS2 meningitis and its molecular mechanisms remain unclear. Epidermal growth factor receptor (EGFR) has been reported to initiate transduction of intracellular signals and regulate host inflammatory responses. Whether and how EGFR contributes to the development of S. suis meningitis are currently unknown. METHODS: The tyrosine phosphorylation of cellular proteins, the transactivation of EGFR, as well as its dimerization, and the associated signal transduction pathways were investigated by immunoprecipitation and western blotting. Real-time quantitative PCR was used to investigate the transcriptional level of the ErbB family members, EGFR-related ligands, cytokines, and chemokines. The secretion of cytokines and chemokines in the serum and brain were detected by Q-Plex™ Chemiluminescent ELISA. RESULTS: We found an important role of EGFR in SS2 strain SC19-induced meningitis. SC19 increasingly adhered to human brain microvascular endothelial cells (hBMEC) and caused inflammatory lesions in the brain tissues, with significant induction and secretion of proinflammatory cytokines and chemokines in the serum and brains. SC19 infection of hBMEC induced tyrosine phosphorylation of cellular EGFR in a ligand-dependent manner involving the EGF-like ligand HB-EGF, amphiregulin (AREG), and epiregulin (EREG) and led to heterodimerization of EGFR/ErbB3. The EGFR transactivation did not participate in SS2 strain SC19 adhesion of hBMEC, as well as in bacterial colonization in vivo. However, its transactivation contributed to the bacterial-induced neuroinflammation, via triggering the MAPK-ERK1/2 and NF-κB signaling pathways in hBMEC that promote the production of proinflammatory cytokines and chemokines. CONCLUSIONS: We investigated for the first time the tyrosine phosphorylation of cellular proteins in response to SS2 strain SC19 infection of hBMEC and demonstrated the contribution of EGFR to SS2-induced neuroinflammation. These observations propose a novel mechanism involving EGFR in SS2-mediated inflammatory responses in the brain, and therefore, EGFR might be an important host target for further investigation and prevention of neuroinflammation caused by SS2 strains.


Assuntos
Encéfalo/metabolismo , Receptores ErbB/metabolismo , Meningite , Infecções Estreptocócicas/complicações , Infecções Estreptocócicas/fisiopatologia , Streptococcus suis/fisiologia , Ativação Transcricional/fisiologia , Anfirregulina/metabolismo , Animais , Encéfalo/microbiologia , Encéfalo/patologia , Citocinas/genética , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Inibidores Enzimáticos/farmacologia , Receptores ErbB/genética , Feminino , Humanos , Meningite/etiologia , Meningite/microbiologia , Meningite/fisiopatologia , Camundongos , Fosforilação/efeitos dos fármacos , Quinazolinas/farmacologia , Receptor ErbB-3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Infecções Estreptocócicas/microbiologia , Suínos , Tirosina/metabolismo , Tirfostinas/farmacologia
14.
Pol J Microbiol ; 65(1): 105-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27282001

RESUMO

The aim of this study was to investigate the capacity of Streptococcus suis strains to form biofilms and to evaluate the antimicrobial activity of Penicillin G and N-acetylcystein (NAC) on both S. suis sessile and planktonic forms. Only non-typeable isolates of S. suis were correlated with a greater biofilm formation capacity. The MCI of Penicillin G and NAC required for inhibiting biofilm growth were higher than the required concentration for inhibiting planktonic growth. The combinations of NAC and Penicillin G showed a strong synergistic activity that inhibited biofilm formation and disrupted the pre-formed biofilm of S. suis.


Assuntos
Acetilcisteína/farmacologia , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Penicilina G/farmacologia , Streptococcus suis/efeitos dos fármacos , Streptococcus suis/fisiologia , Biofilmes/crescimento & desenvolvimento
15.
Sci Rep ; 6: 21241, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26883762

RESUMO

The main role of CodY, a global regulatory protein in most low G + C gram-positive bacteria, is in transcriptional repression. To study the functions of CodY in Streptococcus suis serotype 2 (S. suis 2), a mutant codY clone named ∆codY was constructed to explore the phenotypic variation between ∆codY and the wild-type strain. The result showed that the codY mutation significantly inhibited cell growth, adherence and invasion ability of S. suis 2 to HEp-2 cells. The codY mutation led to decreased binding of the pathogen to the host cells, easier clearance by RAW264.7 macrophages and decreased growth ability in fresh blood of Cavia porcellus. The codY mutation also attenuated the virulence of S. suis 2 in BALB/c mice. Morphological analysis revealed that the codY mutation decreased the thickness of the capsule of S. suis 2 and changed the surface structures analylized by SDS-PAGE. Finally, the codY mutation altered the expressions of many virulence related genes, including sialic acid synthesis genes, leading to a decreased sialic acid content in capsule. Overall, mutation of codY modulated bacterial virulence by affecting the growth and colonization of S. suis 2, and at least via regulating sialic acid synthesis and capsule thickness.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Repressoras/metabolismo , Streptococcus suis/fisiologia , Animais , Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Linhagem Celular , Modelos Animais de Doenças , Feminino , Regulação Bacteriana da Expressão Gênica , Hemólise , Macrófagos/microbiologia , Macrófagos/fisiologia , Camundongos , Viabilidade Microbiana/genética , Viabilidade Microbiana/imunologia , Mutação , Fagocitose/genética , Fagocitose/imunologia , Proteínas Repressoras/genética , Sorogrupo , Infecções Estreptocócicas/microbiologia , Streptococcus suis/classificação , Streptococcus suis/isolamento & purificação , Streptococcus suis/patogenicidade , Suínos , Virulência/genética
16.
Artigo em Inglês | MEDLINE | ID: mdl-26636044

RESUMO

Bacterial cell wall (CW) and extracellular (EC) proteins are often involved in interactions with extracellular matrix (ECM) proteins such as laminin (LN) and fibronectin (FN), which play important roles in adhesion and invasion. In this study, an efficient method combining proteomic analysis and Far-Western blotting assays was developed to screen directly for bacterial surface proteins with LN- and FN-binding capacity. With this approach, fifteen potential LN-binding proteins and five potential FN-binding proteins were identified from Streptococcus suis serotype 2 (SS2) CW and EC proteins. Nine newly identified proteins, including oligopeptide-binding protein OppA precursor (OppA), elongation factor Tu (EF-Tu), enolase, lactate dehydrogenase (LDH), fructose-bisphosphate aldolase (FBA), 3-ketoacyl-ACP reductase (KAR), Gly ceraldehyde-3-phosphate dehydrogenase (GAPDH), Inosine 5'-monophosphate dehydrogenase (IMPDH), and amino acid ABC transporter permease (ABC) were cloned, expressed, purified and further confirmed by Far-Western blotting and ELISA. Five proteins (OppA, EF-Tu, enolase, LDH, and FBA) exhibited specifically binding activity to both human LN and human FN. Furthermore, seven important recombinant proteins were selected and identified to have the ability to bind Hep-2 cells by the indirect immunofluorescent assay. In addition, four recombinant proteins, and their corresponding polyclonal antibodies, were observed to decrease SS2 adhesion to Hep-2 cells, which indicates that these proteins contribute to the adherence of SS2 to host cell surface. Collectively, these results show that the approach described here represents a useful tool for investigating the host-pathogen interactions.


Assuntos
Adesinas Bacterianas/metabolismo , Far-Western Blotting/métodos , Fibronectinas/metabolismo , Laminina/metabolismo , Streptococcus suis/fisiologia , Linhagem Celular , Células Epiteliais/metabolismo , Humanos , Ligação Proteica , Proteômica/métodos
17.
Microb Pathog ; 89: 128-39, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26427882

RESUMO

Pentraxin 3 (PTX3), a soluble pattern recognition receptor, plays an important role in innate immunity and has been implicated to be a candidate resistance gene against Streptococcus suis 2 infection. To discover the antibacterial effect of porcine PTX3 against S. suis 2, the 42-kDa PTX3 protein was expressed by Chinese hamster ovary cells (CHO), and an additional eukaryotic expression vector pVAX-ptx3 was constructed. The expressed porcine PTX3 mediated a range of antibacterial activities including increasing phagocytic capacity of primary porcine alveolar macrophages (PAM) against S. suis 2 and inhibiting adhesion of S. suis 2 to human epidermoid cancer cells (Hep-2). In mouse model, pre-intramuscular injecting with pVAX-ptx3 reduced mortality and reduced bacteria loads in blood, spleen, lung and brain compared with that of control group during 2-12 h following intraperitoneal injection (i.p.) with S. suis 2. Meanwhile, the expressions of IL-6 and IL-8 in blood were increased in pVAX-ptx3 group, whereas no obvious changes about IL-10. In piglet model, bacteria load in blood of pVAX-ptx3 group was significantly lower than that of control group after i.p. with S. suis 2, correspondingly, expression of IL-6 and IL-8 were significantly increased in pVAX-ptx3 group. In contrast, white blood cell (WBC) and neutrophil cell (NEU) count of peripheral blood in pVAX-ptx3 group were lower than that of control group. These studies described a novel antibacterial role for porcine PTX3 against S. suis 2 both in vitro and in vivo and suggested that porcine PTX3 may be a potential biological agent against S. suis 2 in pig and be used for the clinical prevention and treatment of streptococcosis caused by S. suis 2.


Assuntos
Proteína C-Reativa/farmacologia , Fatores Imunológicos/farmacologia , Componente Amiloide P Sérico/farmacologia , Streptococcus suis/efeitos dos fármacos , Estruturas Animais/microbiologia , Animais , Aderência Bacteriana/efeitos dos fármacos , Carga Bacteriana , Proteína C-Reativa/administração & dosagem , Proteína C-Reativa/genética , Proteína C-Reativa/isolamento & purificação , Células Epiteliais/microbiologia , Fatores Imunológicos/administração & dosagem , Injeções Intramusculares , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Camundongos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Componente Amiloide P Sérico/administração & dosagem , Componente Amiloide P Sérico/genética , Componente Amiloide P Sérico/isolamento & purificação , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/prevenção & controle , Streptococcus suis/imunologia , Streptococcus suis/fisiologia , Análise de Sobrevida , Suínos
18.
J Microbiol Biotechnol ; 25(6): 771-81, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25537722

RESUMO

To examine if the molecular chaperone DnaK operon proteins of Streptococcus suis type 2 (SS2) are involved in adhesion to host cells, the abundance values of these proteins from the surface of two SS2 strains of different adhesion capability were compared. Their roles in growth and adhesion to human laryngeal epithelial cell line HEp-2 cells were investigated on SS2 strain HA9801 and its mutants with DnaK operon genes partially knocked-out (PKO mutant) under heat stress. The major difference was that DnaJ was more abundant in strain HA9801 than in strain JX0811. Pretreatment of the bacteria with hyperimmune sera to DnaJ, but not with those to other proteins, could significantly reduce SS2 adhesion to HEp-2 cells. PKO of dnaJ g ene resulted in decreased SS2 growth at 37 °C and 42 °C, and reduced its adhesion to HEp-2 cells. The wild-type strain stressed at 42 °C had increased expression of DnaJ on its surface and elevated adhesion to HEp-2 cells, which was also inhibitable by DnaJ specific antiserum. These results indicate that the DnaJ of S. suis type 2 is important not only for thermotolerance but also for adhesion to host cells. Because DnaJ expression is increased upon temperature upshift with increased exposure on the bacterial surface, the febrile conditions of the cases with systemic infections might help facilitate bacterial adhesion to host cells. DnaJ could be one of the potential candidates as a subunit vaccine because of its good immunogenicity.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Proteínas de Choque Térmico HSP40/metabolismo , Streptococcus suis/enzimologia , Streptococcus suis/fisiologia , Estresse Fisiológico , Proteínas de Bactérias/genética , Linhagem Celular , Células Epiteliais/microbiologia , Técnicas de Inativação de Genes , Proteínas de Choque Térmico HSP40/genética , Temperatura Alta , Humanos , Streptococcus suis/genética , Streptococcus suis/efeitos da radiação
19.
J Infect Dis ; 212(1): 95-105, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25525050

RESUMO

BACKGROUND: Streptococcus suis has emerged as an important cause of bacterial meningitis in adults. The ingestion of undercooked pork is a risk factor for human S. suis serotype 2 (SS2) infection. Here we provide experimental evidence indicating that the gastrointestinal tract is an entry site of SS2 infection. METHODS: We developed a noninvasive in vivo model to study oral SS2 infection in piglets. We compared in vitro interaction of S. suis with human and porcine intestinal epithelial cells (IEC). RESULTS: Two out of 15 piglets showed clinical symptoms compatible with S. suis infection 24-48 hours after ingestion of SS2. SS2 was detected in mesenteric lymph nodes of 40% of challenged piglets. SS2 strains isolated from patients showed significantly higher adhesion to human IEC compared to invasive strains isolated from pigs. In contrast, invasive SS9 strains showed significantly higher adhesion to porcine IEC. Translocation across human IEC, which occurred predominately via a paracellular route, was significantly associated with clonal complex 1, the predominant zoonotic genotype. Adhesion and translocation were dependent on capsular polysaccharide production. CONCLUSIONS: SS2 should be considered a food-borne pathogen. S. suis interaction with human and pig IEC correlates with S. suis serotype and genotype, which can explain the zoonotic potential of SS2.


Assuntos
Interações Hospedeiro-Patógeno , Mucosa Intestinal/microbiologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/veterinária , Streptococcus suis/fisiologia , Zoonoses/microbiologia , Adulto , Animais , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Humanos , Masculino , Meningites Bacterianas/microbiologia , Meningites Bacterianas/veterinária , Suínos
20.
Appl Environ Microbiol ; 81(3): 976-85, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25416757

RESUMO

Streptococcus suis is an emerging zoonotic pathogen causing severe infections in pigs and humans. In previous studies, 33 serotypes of S. suis have been identified using serum agglutination. Here, we describe a novel S. suis strain, CZ130302, isolated from an outbreak of acute piglet meningitis in eastern China. Strong pathogenicity of meningitis caused by strain CZ130302 was reproduced in the BALB/c mouse model. The strain showed a high fatality rate (8/10), higher than those for known virulent serotype 2 strains P1/7 (1/10) and 9801 (2/10). Cell adhesion assay results with bEnd.3 and HEp2 cells showed that CZ130302 was significantly close to P1/7 and 9801. Both the agglutination test and its complementary test showed that strain CZ130302 had no strong cross-reaction with the other 33 S. suis serotypes. The multiplex PCR assays revealed no specified bands for all four sets used to detect the other 33 serotypes. In addition, genetic analysis of the whole cps gene clusters of all serotypes was performed in this study. The results of comparative genomics showed that the cps gene cluster of CZ130302, which was not previously reported, showed no homology to the gene sequences of the other strains. Especially, the wzy, wzx, and acetyltransferase genes of strain CZ130302 are phylogenetically distinct from strains of the other 33 serotypes. Therefore, this study suggested that strain CZ130302 represents a novel variant serotype of S. suis (designated serotype Chz) which has a high potential to be virulent and associated with meningitis in animals.


Assuntos
Meningite/veterinária , Infecções Estreptocócicas/veterinária , Streptococcus suis/classificação , Streptococcus suis/isolamento & purificação , Doenças dos Suínos/microbiologia , Animais , Adesão Celular , China , DNA Bacteriano/química , DNA Bacteriano/genética , Modelos Animais de Doenças , Testes de Hemaglutinação , Células Hep G2 , Hepatócitos/microbiologia , Humanos , Meningite/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Multiplex , Análise de Sequência de DNA , Homologia de Sequência , Sorogrupo , Infecções Estreptocócicas/microbiologia , Streptococcus suis/genética , Streptococcus suis/fisiologia , Análise de Sobrevida , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA