Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Basic Microbiol ; 64(9): e2400030, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39031597

RESUMEN

Streptococcus suis is an important zoonotic pathogen, causing cytokine storms of Streptococcal toxic shock-like syndrome amongst humans after a wound infection into the bloodstream. To overcome the challenges of fever and leukocyte recruitment, invasive S. suis must deploy multiple stress responses forming a network and utilize proteases to degrade short-lived regulatory and misfolded proteins induced by adverse stresses, thereby adapting and evading host immune responses. In this study, we found that S. suis encodes multiple ATP-dependent proteases, including single-chain FtsH and double-subunit Clp protease complexes ClpAP, ClpBP, ClpCP, and ClpXP, which were activated as the fever of infected mice in vivo. The expression of genes ftsH, clpA/B/C, and clpP, but not clpX, were significantly upregulated in S. suis in response to heat stress, while were not changed notably under the treatments with several other stresses, including oxidative, acidic, and cold stimulation. FtsH and ClpP were required for S. suis survival within host blood under heat stress in vitro and in vivo. Deletion of ftsH or clpP attenuated the tolerance of S. suis to heat, oxidative and acidic stresses, and significantly impaired the bacterial survival within macrophages. Further analysis identified that repressor CtsR directly binds and controls the clpA/B/C and clpP operons and is relieved by heat stress. In summary, the deployments of multiple ATP-dependent proteases form a flexible heat stress response network that appears to allow S. suis to fine-tune the degradation or refolding of the misfolded proteins to maintain cellular homeostasis and optimal survival during infection.


Asunto(s)
Proteínas Bacterianas , Infecciones Estreptocócicas , Streptococcus suis , Streptococcus suis/enzimología , Streptococcus suis/genética , Streptococcus suis/patogenicidad , Animales , Ratones , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Infecciones Estreptocócicas/microbiología , Respuesta al Choque Térmico , Proteasas ATP-Dependientes/metabolismo , Proteasas ATP-Dependientes/genética , Regulación Bacteriana de la Expresión Génica , Macrófagos/microbiología , Macrófagos/inmunología , Endopeptidasa Clp/metabolismo , Endopeptidasa Clp/genética , Femenino
2.
BMC Vet Res ; 20(1): 337, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39080654

RESUMEN

Streptococcus suis (S. suis) is an important gram-positive pathogen and an emerging zoonotic pathogen that causes meningitis in swine and humans. Although several virulence factors have been characterized in S. suis, the underlying mechanisms of pathogenesis are not fully understood. In this study, we identified Zinc metalloproteinase C (ZmpC) probably as a critical virulence factor widely distributed in S. suis strains. ZmpC was identified as a critical facilitator in the development of bacterial meningitis, as evidenced by the detection of increased expression of TNF-α, IL-8, and matrix metalloprotease 9 (MMP-9). Subcellular localization analysis further revealed that ZmpC was localized to the cell wall surface and gelatin zymography analysis showed that ZmpC could cleave human MMP-9. Mice challenge demonstrated that ZmpC provided protection against S. suis CZ130302 (serotype Chz) and ZY05719 (serotype 2) infection. In conclusion, these results reveal that ZmpC plays an important role in promoting CZ130302 to cause mouse meningitis and may be a potential candidate for a S. suis CZ130302 vaccine.


Asunto(s)
Meningitis Bacterianas , Serogrupo , Infecciones Estreptocócicas , Streptococcus suis , Enfermedades de los Porcinos , Streptococcus suis/patogenicidad , Streptococcus suis/enzimología , Animales , Infecciones Estreptocócicas/veterinaria , Infecciones Estreptocócicas/microbiología , Porcinos , Enfermedades de los Porcinos/microbiología , Ratones , Meningitis Bacterianas/veterinaria , Meningitis Bacterianas/microbiología , Femenino , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Ratones Endogámicos BALB C , Metaloendopeptidasas/metabolismo , Metaloendopeptidasas/genética
3.
Virulence ; 15(1): 2306719, 2024 12.
Artículo en Inglés | MEDLINE | ID: mdl-38251714

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas , Infecciones Estreptocócicas , Streptococcus suis , Animales , Ratones , Arginina , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Hidrolasas/genética , Hidrolasas/metabolismo , Macrófagos , Infecciones Estreptocócicas/microbiología , Streptococcus suis/patogenicidad , Streptococcus suis/fisiología
4.
Vet Sci ; 10(3)2023 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-36977278

RESUMEN

Streptococcus suis serotype 2 (SS2) is a noteworthy zoonotic pathogen that has been responsible for large economic losses in pig production and a great threat to human health. Pentraxin 3 (PTX3) is an essential regulator of the innate immune response to bacterial pathogens; however, its role during SS2 infection is not fully understood. In this study, we found that the SS2 strain HA9801 induced a significant inflammatory response in the mouse air pouch model; this response was amplified by the treatment of exogenous PTX3 simultaneously in terms of the results of inflammatory cell recruitment and proinflammatory cytokine IL-6 production. In addition, PTX3 facilitated the phagocytosis of macrophage Ana-1 against SS2 strain HA9801. The supplementation of exogenous PTX3 significantly reduced the bacterial loads in a dose-dependent manner in lungs, livers and bloods of SS2-infected mice compared to the samples with HA9801 infection alone; this finding indicated that PTX3 may facilitate the bacterial clearance through enhancing the host inflammatory response during SS2 infection. Both PTX3 and SS2 capsular polysaccharide (CPS2) were required for the robust inflammatory response, implying that the host PTX3 protein and SS2 surface CPS2 modulate the host innate immune response in concert. All of these results suggested that PTX3 is a potential novel biological agent for the SS2 infection; however, the recommended dose of PTX3 must be evaluated strictly to avoid inducing an excessive inflammatory response that can cause serious tissue injury and animal death.

5.
Virulence ; 13(1): 1304-1314, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35903019

RESUMEN

Streptococcus suis serotype 2 (SS2), an emerging zoonotic pathogen, causes swine diseases and human cases of streptococcal toxic shock syndrome. RNA-binding proteins (RBPs) can modulate gene expression through post-transcriptional regulation. In this study, we identified an RBP harbouring an S1 domain, named RbpA, which facilitated SS2 adhesion to host epithelial cells and contributed to bacterial pathogenicity. Comparative proteomic analysis identified 145 proteins that were expressed differentially between ΔrbpA strain and wild-type strain, including several virulence-associated factors, such as the extracellular protein factor (EF), SrtF pilus, IgA1 protease, SBP2 pilus, and peptidoglycan-binding LysM' proteins. The mechanisms underlying the regulatory effects of RbpA on their encoding genes were explored, and it was found that RbpA regulates gene expression through diverse mechanisms, including post-transcriptional regulation, and thus acts as a global regulator. These results partly reveal the pathogenic mechanism mediated by RbpA, improving our understanding of the regulatory systems of S. suis and providing new insights into bacterial pathogenicity.


Asunto(s)
Infecciones Estreptocócicas , Streptococcus suis , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Humanos , Proteómica , Motivos de Unión al ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Serogrupo , Infecciones Estreptocócicas/microbiología , Porcinos , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
6.
Front Microbiol ; 12: 671706, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34475853

RESUMEN

Toxin-antitoxin (TA) systems are ubiquitous genetic elements that play an essential role in multidrug tolerance and virulence of bacteria. So far, little is known about the TA systems in Streptococcus suis. In this study, the Xress-MNTss TA system, composed of the MNTss toxin in the periplasmic space and its interacting Xress antitoxin, was identified in S. suis. ß-galactosidase activity and electrophoretic mobility shift assay (EMSA) revealed that Xress and the Xress-MNTss complex could bind directly to the Xress-MNTss promoter as well as downregulate streptomycin adenylyltransferase ZY05719_RS04610. Interestingly, the Xress deletion mutant was less pathogenic in vivo following a challenge in mice. Transmission electron microscopy and adhesion assays pointed to a significantly thinner capsule but greater biofilm-formation capacity in ΔXress than in the wild-type strain. These results indicate that Xress-MNTss, a new type II TA system, plays an important role in antibiotic resistance and pathogenicity in S. suis.

7.
Vet Res ; 52(1): 50, 2021 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-33762005

RESUMEN

Streptococcus suis (S. suis) is an important zoonotic pathogen that causes septicaemia, meningitis and streptococcal toxic shock-like syndrome in its host, and recent studies have shown that S. suis could be competent for natural genetic transformation. Transformation is an important mechanism for the horizontal transfer of DNA, but some elements that affect the transformation process need to be further explored. Upon entering the competent state, Streptococcus species stimulate the transcription of competence-related genes that are responsible for exogenous DNA binding, uptake and processing. In this study, we performed conserved promoter motif and qRT-PCR analyses and identified CrfP as a novel murein hydrolase that is widespread in S. suis and stimulated with a peptide pheromone in the competent state through a process controlled by ComX. A bioinformatics analysis revealed that CrfP consists of a CHAP hydrolase domain and two bacterial Src homology 3-binding (SH3b) domains. Further characterization showed that CrfP could be exported to extracellular bacterial cells and lytic S. suis strains of different serotypes, and this finding was verified by TEM and a turbidity assay. To investigate the potential effect of CrfP in vivo, a gene-deletion mutant (ΔcrfP) was constructed. Instead of stopping the natural transformation process, the inactivation of CrfP clearly reduced the effective transformation rate. Overall, these findings provide evidence showing that CrfP is important for S. suis serovar 2 competence.


Asunto(s)
Proteínas Bacterianas/genética , Hidrolasas/genética , Infecciones Estreptocócicas/veterinaria , Streptococcus suis/genética , Enfermedades de los Porcinos/microbiología , Animales , Proteínas Bacterianas/metabolismo , Eliminación de Gen , Hidrolasas/metabolismo , Serogrupo , Infecciones Estreptocócicas/microbiología , Streptococcus suis/enzimología , Sus scrofa , Porcinos , Transformación Bacteriana
8.
Transbound Emerg Dis ; 68(4): 2005-2016, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32969570

RESUMEN

Porcine extraintestinal pathogenic Escherichia coli (ExPEC) is occurring with increasing frequency in China, which causes acute septicemia and sudden death in pigs leading to significant economic losses. Bacterial survival and even proliferation within host bloodstream are a common manifestation of a number of bacterial septicemias, including porcine ExPEC diseases. However, the underlying pathogenesis for this novel pathotype of ExPEC has not been explored deeply. Here, we used a conjunction with transposon mutagenesis to identify the mechanisms of bacterial fitness involved in optimal growth of porcine ExPEC in swine serum ex vivo under static culture. Our work identified 28 genes involved in nucleotide biosynthesis, extracellular polysaccharide biosynthesis, regulators Fur and FNR, acid/zinc resistance, and Deley-Douderoff carbon metabolism that are required for the serum fitness. Subsequent functional analyses revealed that either interruption of de novo nucleotide biosynthesis or blocking of several extracellular polysaccharide biosynthesis including O2-antigen, Lipid A-core, and ECA significantly affect porcine ExPEC's growth in swine serum and proliferation in host bloodstream. Furthermore, the reasonable regulations of iron and anaerobic metabolisms in response to host stimuli by global regulators Fur and FNR also play key roles during systemic infection of porcine ExPEC. These findings provide compelling evidences that de novo nucleotide biosynthesis may enable porcine ExPEC to adapt to swine blood-specific nutrient availability, and the effective assembly of O-antigen, lipid A-core, and ECA is required to resist the bactericidal activity of swine serum. These studies contribute to better understand the underlying mechanisms employed by porcine ExPEC to survive, grow in the swine bloodstream, and cause disease. These related factors may serve as therapeutic targets for countering or preventing ExPEC serum resistance in the clinic.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli Patógena Extraintestinal , Enfermedades de los Porcinos , Animales , China , Infecciones por Escherichia coli/veterinaria , Proteínas de Escherichia coli , Escherichia coli Patógena Extraintestinal/genética , Porcinos , Factores de Virulencia/genética
9.
Virulence ; 11(1): 1539-1556, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33138686

RESUMEN

The Streptococcus suis serotype 2 (SS2) is a significant zoonotic pathogen that is responsible for various swine diseases, even causing cytokine storms of Streptococcal toxic shock-like syndromes amongst human. Cell wall anchoring proteins with a C-terminal LPxTG are considered to play vital roles during SS2 infection; however, their exporting mechanism across cytoplasmic membranes has remained vague. This study found that YSIRK-G/S was involved in the exportation of LPxTG-anchoring virulence factors MRP and SspA in virulent SS2 strain ZY05719. The whole-genome analysis indicated that diverse LPxTG proteins fused with an N-terminal YSIRK-G/S motif are encoded in strain ZY05719. Two novel LPxTG proteins SspB and YzpA were verified to be exported via a putative transport system that was dependent on the YSIRK-G/S directed translocation, and portrayed vital functions during the infection of SS2 strain ZY05719. Instead of exhibiting an inactivation of C5a peptidase in SspB, another LPxTG protein with an N-terminal YSIRK-G/S motif from Streptococcus agalactiae was depicted to cleave the C5a component of the host complement. The consequent domain-architecture retrieval determined more than 10,000 SspB/YzpA like proteins that are extensively distributed in the Gram-positive bacteria, and most of them harbor diverse glycosyl hydrolase or peptidase domains within their middle regions, thus presenting their capability to interact with host cells. The said findings provide compelling evidence that LPxTG proteins with an N-terminal YSIRK-G/S motif are polymorphic effectors secreted by Gram-positive bacteria, which can be further proposed to define as cell wall anchoring effectors in a new subset.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Traslocación Bacteriana/genética , Pared Celular/metabolismo , Streptococcus suis/genética , Streptococcus suis/patogenicidad , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Animales , Endopeptidasas/genética , Endopeptidasas/metabolismo , Femenino , Ratones , Ratones Endogámicos BALB C , Streptococcus suis/metabolismo , Porcinos , Enfermedades de los Porcinos/microbiología , Virulencia
10.
Vet Microbiol ; 249: 108830, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32920372

RESUMEN

Porcine extraintestinal pathogenic Escherichia coli (ExPEC) is occurring with increasing frequency in China, and leads to significant economic and welfare costs in the swine industry. The underlying mechanisms of porcine ExPEC in blood colonization during systematic infection is poorly understood. Here we measured the gene expression of porcine ExPEC in infected animal bloodstream in vivo and fresh swine blood in vitro. Using comparisons with P values of ≤ 0.01, we identified 354 and 313 genes as being significantly up- or down-regulated at least 2-fold change during bloodstream infection, respectively. Excepting for an array of iron acquisition systems, numerous genes involved in carbon central metabolism and anaerobic respiratory chains were upregulated here. These genes were categorized into several clusters including the TCA-cycle (frdABCD, citCEFXG), d-ribose transporter (rbsDACB), nickel transporter (nikABCDER), NiFe hydrogenase (hybOABCDEF, hycBCDEFG), Hyp-complex (hypABCDE), DMSO reductase (dmsABC and ynfEFGHI), format dehydrogenase (fdnGHI) and NADH dehydrogenase I (nuoA-N). The mutant with simultaneous inactivation of ribose and citrate imports showed significant reduced fitness in host blood, suggesting these two carbohydrates are utilized by central metabolism network as important carbon-source during bloodstream infection. Similar deficiency was also observed in the mutant double deleted NiFe hydrogenase 2 and 3 anaerobic respiratory chains. Further study found that FNR (a global regulator facilitating bacterial adaptation to anaerobic conditions) is an important regulator in response to bloodstream to activate center metabolism and anaerobic respiratory chains, thus contribute to the full-virulence of porcine ExPEC. These findings provide compelling evidence to support the notion that carbon central metabolism network and anaerobic respiratory chains play key roles for porcine ExPEC fitness within host bloodstream.


Asunto(s)
Bacteriemia/veterinaria , Infecciones por Escherichia coli/veterinaria , Proteínas de Escherichia coli/genética , Escherichia coli Patógena Extraintestinal/genética , Regulación Bacteriana de la Expresión Génica , Enfermedades de los Porcinos/microbiología , Anaerobiosis/genética , Animales , Bacteriemia/microbiología , Carbono/metabolismo , China , Transporte de Electrón/genética , Infecciones por Escherichia coli/microbiología , Escherichia coli Patógena Extraintestinal/metabolismo , Porcinos
11.
Pathogens ; 9(5)2020 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-32443590

RESUMEN

Infection with the epidemic virulent strain of Streptococcus suis serotype 2 (SS2) can cause septicemia in swine and humans, leading to pneumonia, meningitis and even cytokine storm of Streptococcal toxic shock-like syndrome. Despite some progress concerning the contribution of bacterial adhesion, biofilm, toxicity and stress response to the SS2 systemic infection, the precise mechanism underlying bacterial survival and growth within the host bloodstream remains elusive. Here, we reported the SS2 virulent strains with a more than 20 kb endoSS-related insertion region that showed significantly higher proliferative ability in swine serum than low-virulent strains. Further study identified a complete N-glycans degradation system encoded within this insertion region, and found that both GH92 and EndoSS contribute to bacterial virulence, but that only DndoSS was required for optimal growth of SS2 in host serum. The supplement of hydrolyzed high-mannose-containing glycoprotein by GH92 and EndoSS could completely restore the growth deficiency of endoSS deletion mutant in swine serum. EndoSS only hydrolyzed a part of the model glycoprotein RNase B with high-mannose N-linked glycoforms into a low molecular weight form, and the solo activity of GH92 could not show any changes comparing with the blank control in SDS-PAGE gel. However, complete hydrolyzation was observed under the co-incubation of EndoSS and GH92, suggesting GH92 may degrade the high-mannose arms of N-glycans to generate a substrate for EndoSS. In summary, these findings provide compelling evidences that EndoSS-related N-glycans degradation system may enable SS2 to adapt to host serum-specific availability of carbon sources from glycoforms, and be required for optimal colonization and full virulence during systemic infection.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA