Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.839
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Neonatal Perinatal Med ; 17(2): 265-268, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38669486

RESUMO

 Streptococcus gallolyticus subspecies pasteurianus is a subtype of Streptococcus bovis (S. bovis) that has become increasingly recognized as a sepsis-causing pathogen in neonates. It is well documented that S. bovis species have a predilection to both cardiac and gastrointestinal tissue, and in adult populations, isolating these organisms in the bloodstream often triggers further evaluation for co-morbid complications such as colon cancer or endocarditis. However, no such guidance currently exists in neonatal literature. We present a case of a preterm infant with S. gallolyticus subsp. pasteurianus bacteremia presenting as necrotizing enterocolitis (NEC) not previously described in the literature. Furthermore, through a complete diagnostic evaluation, including an echocardiogram, our patient was found to have the rare complication of endocarditis.


Assuntos
Enterocolite Necrosante , Recém-Nascido Prematuro , Infecções Estreptocócicas , Humanos , Enterocolite Necrosante/microbiologia , Recém-Nascido , Infecções Estreptocócicas/complicações , Infecções Estreptocócicas/microbiologia , Arterite/microbiologia , Streptococcus gallolyticus subspecies gallolyticus , Masculino , Bacteriemia/microbiologia , Doenças do Prematuro/microbiologia , Feminino , Antibacterianos/uso terapêutico
2.
PLoS One ; 19(4): e0299691, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38568909

RESUMO

Streptococcus suis serotype 2 (SS2) is an important porcine pathogen that causes diseases in both swine and human. For rapid SS2 identification, a novel latex agglutination test (LAT) based on heavy-chain variable domain antibody (VH) was developed. Firstly, the soluble 47B3 VH antibody fragment from a phage display library, in which cysteine residues were engineered at the C-terminus, was expressed in Escherichia coli. The purified protein was then gently reduced to form monomeric soluble 47B3 VH subsequently used to coat with latex beads by means of site-specific conjugation. The resulting VH-coated beads gave a good agglutination reaction with SS2. The LAT was able to distinguish S. suis serotype 2 from serotype 1/2, which shares some common sugar residues, and showed no cross-reaction with other serotypes of S. suis or other related bacteria. The detection sensitivity was found to be as high as 1.85x106 cells. The LAT was stable at 4°C for at least six months without loss of activity. To the best of our knowledge, this is the first LAT based on a VH antibody fragment that can be considered as an alternative for conventional antibody-based LAT where VHs are the most favored recombinant antibody.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Doenças dos Suínos , Animais , Humanos , Suínos , Sorogrupo , Testes de Fixação do Látex/métodos , Fragmentos de Imunoglobulinas , Proteínas Recombinantes/genética , Escherichia coli/genética , Infecções Estreptocócicas/microbiologia , Doenças dos Suínos/microbiologia
3.
Microbiol Spectr ; 12(5): e0426023, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38587390

RESUMO

Streptococcus pyogenes causes a variety of human infections, and hospital outbreaks with this pathogen have also been reported. The purpose of this study is to describe the clinical characteristics of an outbreak of S. pyogenes involving 15 patients and four healthcare workers (HCWs), as well as the molecular characteristics of the causative isolates. The course and response to the outbreak were reviewed, and information on the characteristics of the patients was extracted retrospectively from the medical records. Whole-genome sequencing of the 16 causative isolates (14 from patients and two from HCWs) was also performed. All 15 patients were postoperative of head and neck cancer with tracheotomy, and 12 had invasive infections, primarily surgical site infections, all of which resolved without causing serious illness. All but the first case was detected more than 7 days after admission. S. pyogenes was detected in two patients after empiric antimicrobial administration was performed on all inpatients and HCWs, and the outbreak was finally contained in approximately 2 months. All isolates detected in patients and HCWs belonged to emm89/clade 3, a hypervirulent clone that has emerged worldwide and was classified as sequence type 646. These isolates had single nucleotide polymorphism (SNP) differences of zero to one, indicating clonal transmission. This study demonstrated an outbreak of S. pyogenes emm89/clade 3 in a ward of patients with head and neck cancer. The global emergence of hypervirulent isolates may increase the risk of outbreaks among high-risk patients. IMPORTANCE: This study describes an outbreak of Streptococcus pyogenes that occurred in a ward caring for patients with head and neck cancer and tracheostomies. Many cases of invasive infections occurred in a short period, and extensive empiric antimicrobial administration on patients and healthcare workers was performed to control the outbreak. Whole-genome sequencing analysis of the causative strains confirmed that it was a monoclonal transmission of strains belonging to emm89/clade 3. The epidemiology and clinical characteristics of S. pyogenes infections have changed with the replacement of the prevalent clones worldwide. In the 1980s, there was a reemergence of S. pyogenes infections in high-income countries due to the spread of hypervirulent emm1 strains. emm89/clade 3 has recently been spreading worldwide and shares common features with emm1, including increased production of two toxins, NADase, and streptolysin O. The outbreak reported here may reflect the high spreading potential and virulence of emm89/clade 3.


Assuntos
Infecção Hospitalar , Surtos de Doenças , Neoplasias de Cabeça e Pescoço , Infecções Estreptocócicas , Streptococcus pyogenes , Humanos , Streptococcus pyogenes/genética , Streptococcus pyogenes/isolamento & purificação , Streptococcus pyogenes/classificação , Streptococcus pyogenes/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/microbiologia , Neoplasias de Cabeça e Pescoço/cirurgia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/epidemiologia , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Infecção Hospitalar/epidemiologia , Infecção Hospitalar/microbiologia , Estudos Retrospectivos , Sequenciamento Completo do Genoma , Adulto , Polimorfismo de Nucleotídeo Único , Infecção da Ferida Cirúrgica/microbiologia , Infecção da Ferida Cirúrgica/epidemiologia , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Idoso de 80 Anos ou mais , Pessoal de Saúde/estatística & dados numéricos
4.
Emerg Microbes Infect ; 13(1): 2339946, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38578304

RESUMO

Streptococcus suis is a significant and emerging zoonotic pathogen. ST1 and ST7 strains are the primary agents responsible for S. suis human infections in China, including the Guangxi Zhuang Autonomous Region (GX). To enhance our understanding of S. suis ST1 population characteristics, we conducted an investigation into the phylogenetic structure, genomic features, and virulence levels of 73 S. suis ST1 human strains from GX between 2005 and 2020. The ST1 GX strains were categorized into three lineages in phylogenetic analysis. Sub-lineage 3-1a exhibited a closer phylogenetic relationship with the ST7 epidemic strain SC84. The strains from lineage 3 predominantly harboured 89K-like pathogenicity islands (PAIs) which were categorized into four clades based on sequence alignment. The acquirement of 89K-like PAIs increased the antibiotic resistance and pathogenicity of corresponding transconjugants. We observed significant diversity in virulence levels among the 37 representative ST1 GX strains, that were classified as follows: epidemic (E)/highly virulent (HV) (32.4%, 12/37), virulent plus (V+) (29.7%, 11/37), virulent (V) (18.9%, 7/37), and lowly virulent (LV) (18.9%, 7/37) strains based on survival curves and mortality rates at different time points in C57BL/6 mice following infection. The E/HV strains were characterized by the overproduction of tumour necrosis factor (TNF)-α in serum and promptly established infection at the early phase of infection. Our research offers novel insights into the population structure, evolution, genomic features, and pathogenicity of ST1 strains. Our data also indicates the importance of establishing a scheme for characterizing and subtyping the virulence levels of S. suis strains.


Assuntos
Genoma Bacteriano , Ilhas Genômicas , Filogenia , Infecções Estreptocócicas , Streptococcus suis , Streptococcus suis/genética , Streptococcus suis/patogenicidade , Streptococcus suis/classificação , Streptococcus suis/isolamento & purificação , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/veterinária , Infecções Estreptocócicas/epidemiologia , China/epidemiologia , Humanos , Virulência , Animais , Camundongos , Feminino , Genômica , Fatores de Virulência/genética
5.
Infect Immun ; 92(4): e0006224, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38514466

RESUMO

Streptococcus agalactiae also named Group B Streptococcus (GBS) is the most significant pathogen causing invasive infections, such as bacteremia and meningitis, in neonates. Worldwide epidemiological studies have shown that a particular clonal complex (CC) of capsular serotype III, the CC17, is strongly associated with meningitis in neonates and is therefore, designated as the hypervirulent clone. Macrophages are a permissive niche for intracellular bacteria of all GBS clones. In this study, we deciphered the specific interaction of GBS CC17 strains with macrophages. Our study revealed that CC17 strains are phagocytosed at a higher rate than GBS non-CC17 strains by human monocytes and macrophages both in cellular models and in primary cells. CC17-enhanced phagocytosis is due to an initial enhanced-attachment step to macrophages mediated by the CC17-specific surface protein HvgA and the PI-2b pilus (Spb1). We showed that two different inhibitors of scavenger receptors (fucoidan and poly(I)) specifically inhibited CC17 adhesion and phagocytosis while not affecting those of non-CC17 strains. Once phagocytosed, both CC17 and non-CC17 strains remained in a LAMP-1 positive vacuole that ultimately fuses with lysosomes where they can survive at similar rates. Finally, both strains displayed a basal egress which occurs independently from actin and microtubule networks. Our findings provide new insights into the interplay between the hypervirulent GBS CC17 and major players of the host's innate immune response. This enhanced adhesion, leading to increased phagocytosis, could reflect a peculiar capacity of the CC17 lineage to subvert the host immune defenses, establish a niche for persistence or disseminate.


Assuntos
Meningite , Infecções Estreptocócicas , Recém-Nascido , Humanos , Streptococcus agalactiae , Infecções Estreptocócicas/microbiologia , Macrófagos , Células Clonais
6.
Int Immunopharmacol ; 131: 111922, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38522137

RESUMO

Streptococcus equi subsp. zooepidemicus (SEZ) is a zoonotic bacterial pathogen that causes life-threatening infections and various diseases such as meningitis, endocarditis and pneumonia. With the use of antibiotics being severely restricted in the international community, an alternative to antibiotics is urgently needed against bacterial. In the present study, the herbal extract magnolol protected mice against SEZ infection, reflected by increased survival rate and reduced bacterial burden. A pro-inflammatory form of cell death occurred in SEZ-infected macrophage. Magnolol downregulated the expression of pyroptosis-related proteins and reduced the formation of cell membrane pores in infected macrophages to suppress the development of subsequent inflammation. We further demonstrated that magnolol directly suppressed SEZ-induced macrophage pyroptosis, which partially protected macrophages from SEZ infection. Our study revealed that magnolol suppressed inflammation and protected mice against SEZ infection, providing a possible treatment for SEZ infection.


Assuntos
Compostos de Bifenilo , Lignanas , Infecções Estreptocócicas , Streptococcus equi , Animais , Camundongos , Streptococcus equi/fisiologia , Piroptose , Macrófagos/microbiologia , Inflamação , Antibacterianos , Infecções Estreptocócicas/tratamento farmacológico , Infecções Estreptocócicas/microbiologia
7.
J Vet Diagn Invest ; 36(3): 473-476, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38520120

RESUMO

Gastrointestinal adenocarcinomas are often reported in South American camelids (SAC). We describe here cases of gastroduodenal adenocarcinoma in an adult alpaca (Vicugna pacos) and a llama (Llama glama); both SACs were anorectic and lethargic before death. At autopsy, a prominent and firm caudal C3-pyloric-duodenal junction with stricture and ulceration was present in both animals, as were hemorrhages in various organs and hydrothorax. Microscopically, scattered nests, cords, and tortuous acini of neoplastic epithelial cells were embedded in desmoplastic stroma and invaded the submucosa and muscle layers of the gastroduodenal junction. The mucosa was necrotic, with gram-negative rods in the alpaca and colonies of gram-positive cocci in the llama. No tumor metastases were observed. The neoplastic cells immunolabeled for pancytokeratin. Escherichia coli was isolated from the alpaca and Streptococcus lutetiensis from the llama; septicemia was the cause of death in both animals. Although adenocarcinomas arising from gastric compartments and intestinal segments have been reported in SACs, adenocarcinoma of the caudal C3-pyloric-duodenal junction has not been reported previously in these species, to our knowledge.


Assuntos
Adenocarcinoma , Camelídeos Americanos , Animais , Adenocarcinoma/veterinária , Adenocarcinoma/patologia , Neoplasias Duodenais/veterinária , Neoplasias Duodenais/patologia , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/veterinária , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Evolução Fatal , Sepse/veterinária , Sepse/microbiologia , Infecções Estreptocócicas/veterinária , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia
8.
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
Infecções Estreptocócicas , Streptococcus suis , Animais , Camundongos , Hidrolases/genética , Hidrolases/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Macrófagos , Arginina , Infecções Estreptocócicas/microbiologia , Regulação Bacteriana da Expressão Gênica
9.
Microbiol Spectr ; 12(2): e0280323, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38230928

RESUMO

Streptococcus suis (S. suis) has been increasingly recognized as a porcine zoonotic pathogen that threatens the health of both pigs and humans. Multidrug-resistant Streptococcus suis is becoming increasingly prevalent, and novel strategies to treat bacterial infections caused by these organisms are desperately needed. In the present study, an untargeted metabolomics analysis showed that the significant decrease in methionine content and the methionine biosynthetic pathway were significantly affected by the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis in drug-resistant S. suis. The addition of L-methionine restored the bactericidal activity of macrolides, doxycycline, and ciprofloxacin on S. suis in vivo and in vitro. Further studies showed that the exogenous addition of methionine affects methionine metabolism by reducing S-adenosylmethionine synthetase activity and the contents of S-adenosylmethionine, S-adenosyl homocysteine, and S-ribose homocysteine. Methionine can decrease the total methylation level and methylesterase activity in multidrug resistant S. suis. The drug transport proteins and efflux pump genes were significantly downregulated in S. suis by exogenous L-methionine. Moreover, the exogenous addition of methionine can reduce the survival of S. suis by affecting oxidative stress and metal starvation in bacteria. Thus, L-methionine may influence the development of resistance in S. suis through methyl metabolism and metal starvation. This study provides a new perspective on the mitigation of drug resistance in S. suis.IMPORTANCEBacterial antibiotic resistance has become a severe threat to human and animal health. Increasing the efficacy of existing antibiotics is a promising strategy against antibiotic resistance. Here, we report that L-methionine enhances the efficacy of macrolides, doxycycline, and ciprofloxacin antibiotics in killing Streptococcus suis, including multidrug-resistant pathogens. We investigated the mechanism of action of exogenous methionine supplementation in restoring macrolides in Streptococcus suis and the role of the methionine cycle pathway on methylation levels, efflux pump genes, oxidative stress, and metal starvation in Streptococcus suis. It provides a theoretical basis for the rational use of macrolides in clinical practice and also identifies a possible target for restoring drug resistance in Streptococcus suis.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Humanos , Animais , Suínos , Streptococcus suis/genética , Macrolídeos/uso terapêutico , Metionina/metabolismo , Metionina/uso terapêutico , Doxiciclina/uso terapêutico , Infecções Estreptocócicas/microbiologia , Antibacterianos/uso terapêutico , Ciprofloxacina , Homocisteína/metabolismo , Homocisteína/uso terapêutico
10.
PLoS One ; 19(1): e0295547, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38206970

RESUMO

Streptococcus uberis is frequently isolated from milk collected from dairy cows with mastitis. According to the host's immunity, bacterial virulence, and their interaction, infection with some strains can induce persistent subclinical inflammation, while infection with others induces severe inflammation and transient mastitis. This study compared the inflammatory response of milk-isolated white blood cells (mWBCs) to persistent and transient S. uberis strains. Quarter milk samples were collected aseptically for bacterial culture from all lactating cows once a week over a 10-week period. A transient and noncapsular strain with a 1-week intramammary infection duration was selected from this herd, while a persistent and capsular S. uberis strain with an intramammary infection longer than 2 months from our previous study was selected based on an identical pulse field gel electrophoresis pattern during the IMI episode. Cellular and molecular responses of mWBCs were tested, and the data were analyzed using repeated analysis of variance. The results showed a higher response in migration, reactive oxygen species generation, and bacterial killing when cells were stimulated with transient S. uberis. In contrast, the persistent strain led to increased neutrophil extracellular trap release. This study also highlighted several important molecular aspects of mWBCs. Gene expression analyses by real-time RT-PCR revealed a significant elevation in the expression of Toll-like receptors (TLR-1, TLR-2, TLR-6) and proinflammatory cytokines (tumor necrosis factor-alpha or TNF-α) with the transient strain. Additionally, Streptococcus uberis capsule formation might contribute to the capability of these strains to induce different immune responses. Altogether, these results focus on the immune function of activated mWBCs which demonstrate that a transient strain can elicit a stronger local immune response and, subsequently, lead to rapid recovery from mastitis.


Assuntos
Mastite Bovina , Infecções Estreptocócicas , Streptococcus , Animais , Feminino , Bovinos , Humanos , Leite/metabolismo , Infecções Estreptocócicas/microbiologia , Lactação , Mastite Bovina/microbiologia , Fagócitos , Inflamação/metabolismo
11.
Intern Med ; 63(3): 457-460, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37344440

RESUMO

Streptococcus salivarius is part of the normal oral cavity and gastrointestinal tract microflora and an unusual cause of acute bacterial meningitis. We herein report an 81-year-old man with S. salivarius meningitis, which led to a diagnosis of early esophageal cancer and early gastric cancer. S. salivarius infection may occur through the gastrointestinal mucosa when it is disrupted in association with early gastrointestinal cancer. To our knowledge, this is the first report describing S. salivarius meningitis associated with multiple early gastrointestinal cancers in the absence of other sources of infection.


Assuntos
Neoplasias Esofágicas , Meningites Bacterianas , Neoplasias Gástricas , Infecções Estreptocócicas , Streptococcus salivarius , Masculino , Humanos , Idoso de 80 Anos ou mais , Infecções Estreptocócicas/complicações , Infecções Estreptocócicas/diagnóstico , Infecções Estreptocócicas/microbiologia , Neoplasias Gástricas/complicações , Neoplasias Gástricas/diagnóstico , Meningites Bacterianas/complicações , Meningites Bacterianas/diagnóstico , Meningites Bacterianas/microbiologia , Neoplasias Esofágicas/complicações
12.
PLoS One ; 18(11): e0294868, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38033043

RESUMO

Streptococcus gallolyticus sp. gallolyticus (SGG) is a gut pathobiont involved in the development of colorectal cancer (CRC). To decipher SGG contribution in tumor initiation and/or acceleration respectively, a global transcriptome was performed in human normal colonic cells (FHC) and in human tumoral colonic cells (HT29). To identify SGG-specific alterations, we chose the phylogenetically closest relative, Streptococcus gallolyticus subsp. macedonicus (SGM) as control bacterium. We show that SGM, a bacterium generally considered as safe, did not induce any transcriptional changes on the two human colonic cells. The transcriptional reprogramming induced by SGG in normal FHC and tumoral HT29 cells was significantly different, although most of the genes up- and down-regulated were associated with cancer disease. Top up-regulated genes related to cancer were: (i) IL-20, CLK1, SORBS2, ERG1, PIM1, SNORD3A for normal FHC cells and (ii) TSLP, BHLHA15, LAMP3, ZNF27B, KRT17, ATF3 for cancerous HT29 cells. The total number of altered genes were much higher in cancerous than in normal colonic cells (2,090 vs 128 genes being affected, respectively). Gene set enrichment analysis reveals that SGG-induced strong ER- (endoplasmic reticulum) stress and UPR- (unfolded protein response) activation in colonic epithelial cells. Our results suggest that SGG induces a pro-tumoral shift in human colonic cells particularly in transformed cells potentially accelerating tumor development in the colon.


Assuntos
Neoplasias Colorretais , Infecções Estreptocócicas , Streptococcus gallolyticus subspecies gallolyticus , Humanos , Neoplasias Colorretais/microbiologia , Streptococcus , Perfilação da Expressão Gênica , Infecções Estreptocócicas/microbiologia , Streptococcus gallolyticus/genética
13.
Sci Rep ; 13(1): 20980, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-38017083

RESUMO

Streptococcus agalactiae (S. agalactiae), group B Streptococcus (GBS), a major cause of infection in a wide variety of diseases, have been compared in different human and animal sources. We aimed to compare the bacterial proteome and metabolome profiles of human and animal S. agalactiae strains to delineate biological interactions relevant to infection. With the innovative advancement in mass spectrometry, a comparative result between both strains provided a solid impression of different responses to the host. For instance, stress-related proteins (Asp23/Gls24 family envelope stress response protein and heat shock protein 70), which play a role in the survival of GBS under extreme environmental conditions or during treatment, are highly expressed in human and animal strains. One human strain contains ꞵ-lactamase (serine hydrolase) and biofilm regulatory protein (lytR), which are important virulence regulators and potential targets for the design of novel antimicrobials. Another human strain contains the aminoglycosides-resistance bifunctional AAC/APH (A0A0U2QMQ5) protein, which confers resistance to almost all clinically used aminoglycosides. Fifteen different metabolites were annotated between the two groups. L-aspartic acid, ureidopropionic acid, adenosine monophosphate, L-tryptophan, and guanosine monophosphate were annotated at higher levels in human strains. Butyric acid, fumaric acid, isoleucine, leucine, and hippuric acid have been found in both human and animal strains. Certain metabolites were uniquely expressed in animal strains, with fold changes greater than 2. For example, putrescine modulates biofilm formation. Overall, this study provides biological insights into the substantial possible bacterial response reflected in its macromolecular production, either at the proteomic or metabolomic level.


Assuntos
Infecções Estreptocócicas , Streptococcus agalactiae , Humanos , Animais , Proteômica , Infecções Estreptocócicas/microbiologia , Virulência , Aminoglicosídeos
14.
Front Cell Infect Microbiol ; 13: 1282622, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37915845

RESUMO

The genus Streptococcus encompasses many bacterial species that are associated with hosts, ranging from asymptomatic colonizers and commensals to pathogens with a significant global health burden. Streptococci produce numerous factors that enable them to occupy their host-associated niches, many of which alter their host environment to the benefit of the bacteria. The ability to manipulate host immune systems to either evade detection and clearance or induce a hyperinflammatory state influences whether bacteria are able to survive and persist in a given environment, while also influencing the propensity of the bacteria to cause disease. Several bacterial factors that contribute to this inter-species interaction have been identified. Recently, small peptides have become increasingly appreciated as factors that contribute to Streptococcal relationships with their hosts. Peptides are utilized by streptococci to modulate their host environment in several ways, including by directly interacting with host factors to disrupt immune system function and signaling to other bacteria to control the expression of genes that contribute to immune modulation. In this review, we discuss the many contributions of Streptococcal peptides in terms of their ability to contribute to pathogenesis and disruption of host immunity. This discussion will highlight the importance of continuing to elucidate the functions of these Streptococcal peptides and pursuing the identification of new peptides that contribute to modulation of host environments. Developing a greater understanding of how bacteria interact with their hosts has the potential to enable the development of techniques to inhibit these peptides as therapeutic approaches against Streptococcal infections.


Assuntos
Interações entre Hospedeiro e Microrganismos , Infecções Estreptocócicas , Humanos , Streptococcus , Infecções Estreptocócicas/microbiologia , Bactérias/metabolismo , Peptídeos/metabolismo
15.
Int J Antimicrob Agents ; 62(6): 106996, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37788717

RESUMO

BACKGROUND: Streptococcus suis is an important zoonotic pathogen that often causes biofilm-associated infection. Bacterial biofilm-dependent infection is associated with enhanced drug resistance, making it difficult to eradicate. Novel therapeutic approaches are required urgently to treat infections associated with S. suis biofilm. This study aimed to investigate the effects and mechanisms of methyl anthranilate (MA) on S. suis biofilm. METHODS: The effect of MA on S. suis biofilm was determined using the crystal violet method, and the microstructure of the biofilm was observed by electron microscopy. The effects on capsular polysaccharides were determined using the phenol-sulphuric acid method and high-performance liquid chromatography. Adhesion and antiphagocytosis properties of S. suis were detected via cell assays. Molecular docking, molecular dynamics simulation and enzyme activity inhibition assays were used to further explore the effect of MA on AI-2 quorum sensing (QS) of S. suis. Finally, the therapeutic effect of MA was investigated using a mouse infection model. RESULTS: MA destroyed the structure of S. suis biofilm, hindered biofilm formation, and reduced the synthesis of capsular polysaccharides significantly, which further weakened the adhesion and antiphagocytosis ability of S. suis. MA had a docking effect and binding site (SER76 and ASP197) similar to S-adenosylhomocysteine (SAH). Further analysis showed that MA competitively bound 5'-methyladenosine/S-adenosine homocysteine nucleosidase with SAH to interfere with AI-2 QS. In a mouse model, MA reduced the bacterial burden and inflammatory infiltrates effectively. CONCLUSION: This study revealed the antibiofilm effects of MA, and highlighted its potential as a QS inhibitor against S. suis infection.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Humanos , Simulação de Acoplamento Molecular , Biofilmes , Polissacarídeos , Infecções Estreptocócicas/tratamento farmacológico , Infecções Estreptocócicas/microbiologia
16.
Infect Immun ; 91(11): e0025823, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37874162

RESUMO

The pro-inflammatory cytokine IL-6 regulates antimicrobial responses that are broadly crucial in the defense against infection. Our prior work shows that IL-6 promotes the killing of the M4 serotype group A Streptococcus (GAS) but does not impact the globally disseminated M1T1 serotype associated with invasive infections. Using in vitro and in vivo infection models, we show that IL-6 induces phagocyte reactive oxygen species (ROS) that are responsible for the differential susceptibility of M4 and M1T1 GAS to IL-6-mediated defenses. Clinical isolates naturally deficient in capsule, or M1T1 strains deficient in capsule production, are sensitive to this ROS killing. The GAS capsule is made of hyaluronic acid, an antioxidant that detoxifies ROS and can protect acapsular M4 GAS when added exogenously. During in vitro interactions with macrophages and neutrophils, acapsular GAS can also be rescued with the antioxidant N-acetylcysteine, suggesting this is a major virulence contribution of the capsule. In an intradermal infection model with gp91phox -/- (chronic granulomatous disease [CGD]) mice, phagocyte ROS production had a modest effect on bacterial proliferation and the cytokine response but significantly limited the size of the bacterial lesion in the skin. These data suggest that the capsule broadly provides enhanced resistance to phagocyte ROS but is not essential for invasive infection. Since capsule-deficient strains are observed across several GAS serotypes and are competent for transmission and both mild and invasive infections, additional host or microbe factors may contribute to ROS detoxification during GAS infections.


Assuntos
Ácido Hialurônico , Infecções Estreptocócicas , Animais , Camundongos , Espécies Reativas de Oxigênio , Antioxidantes , Interleucina-6 , Neutrófilos/microbiologia , Streptococcus pyogenes , Infecções Estreptocócicas/microbiologia , Proteínas de Bactérias
17.
PLoS One ; 18(7): e0288840, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37498866

RESUMO

Streptococcus suis is a zoonotic pathogen that causes invasive infections in humans and pigs. Herein, we performed genomic analysis of seven S. suis serotype 4 strains belonging to clonal complex (CC) 94 that were recovered from a human patient or from diseased and clinically healthy pigs. Genomic exploration and comparisons, as well as in vitro cytotoxicity tests, indicated that S. suis CC94 serotype 4 strains are potentially virulent. Genomic analysis revealed that all seven strains clustered within minimum core genome group 3 (MCG-3) and had a high number of virulence-associated genes similar to those of virulent serotype 2 strains. Cytotoxicity assays showed that both the human lung adenocarcinoma cell line and HeLa cells rapidly lost viability following incubation for 4 h with the strains at a concentration of 106 bacterial cells. The human serotype 4 strain (ID36054) decreased cell viability profoundly and similarly to the control serotype 2 strain P1/7. In addition, strain ST1689 (ID34572), isolated from a clinically healthy pig, presented similar behaviour in an adenocarcinoma cell line and HeLa cells. The antimicrobial resistance genes tet(O) and ermB that confer resistance to tetracyclines, macrolides, and lincosamides were commonly found in the strains. However, aminoglycoside and streptothricin resistance genes were found only in certain strains in this study. Our results indicate that S. suis CC94 serotype 4 strains are potentially pathogenic and virulent and should be monitored.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Doenças dos Suínos , Suínos , Humanos , Animais , Sorogrupo , Virulência/genética , Células HeLa , Genômica , Antibacterianos , Infecções Estreptocócicas/veterinária , Infecções Estreptocócicas/microbiologia , Doenças dos Suínos/microbiologia
18.
Microb Pathog ; 183: 106270, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37499842

RESUMO

Autophagy is a highly conserved cellular defensive mechanism that can eliminate bacterial pathogens such as Streptococcus uberis, that causes mastitis in cows. However, S. uberis induced autophagy is still unclear. In this study, we tested if certain inflammatory cytokines such as IL-6, TNF-α, and IFN-γ, critical in mastitis due to S. uberis infection, regulate autophagy activation in bovine mammary epithelial cells (bMECs). Using Western blot and laser scanning confocal microscope in bMECs challenged by S. uberis, showed that the expression of IL-6, TNF-α, IFN-γ oscillated with the expressions of autophagic Atg5, ULK1, PTEN, P62, and LC3ӀӀ/LC3Ӏ. S. uberis infection induced autophagosomes and LC3 puncta in bMECs with upregulation of Atg5, ULK1, PTEN, LC3ӀӀ/LC3Ӏ, and downregulation of P62. The levels of IL-6, TNF-α, and IFN-γ increased during autophagy flux formation to decrease during autophagy induction. Autophagy inhibition increased the expression of IL-6, TNF-α, and IFN-γ and increased S. uberis burden. This study indicates autophagy is induced during S. uberis infection and IL-6, TNF-α, and IFN-γ contribute to autophagy and autophagy flux formation.


Assuntos
Mastite Bovina , Infecções Estreptocócicas , Feminino , Bovinos , Animais , Humanos , Fator de Necrose Tumoral alfa/metabolismo , Infecções Estreptocócicas/microbiologia , Interleucina-6/metabolismo , Glândulas Mamárias Animais/microbiologia , Interferon gama/metabolismo , Células Epiteliais/microbiologia , Autofagia , Mastite Bovina/microbiologia
19.
Eur J Clin Microbiol Infect Dis ; 42(7): 907-912, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37145237

RESUMO

Whether cirrhotic patients with Streptococcus bovis bacteremia have an increased risk of colorectal neoplasm is uncertain. A multicentric retrospective cohort study was conducted investigating associations between S. bovis biotype and species, cirrhosis, and colorectal neoplasm. Out of 779 patients with S. bovis bacteremia, 69 (8.7%) had cirrhosis. No differences were found in the prevalence of colorectal neoplasm between cirrhotic and non-cirrhotic patients undergoing colonoscopy. Among cirrhotic patients, prevalence of colorectal neoplasms was higher in S. bovis biotype I (S. gallolyticus) bacteremia (80%) than in S. bovis biotype II (33.3%; p < 0.007). In conclusion, risk of colorectal neoplasm is high among cirrhotic patients with S. gallolyticus bacteremia.


Assuntos
Bacteriemia , Neoplasias do Colo , Neoplasias Colorretais , Infecções Estreptocócicas , Streptococcus bovis , Humanos , Estudos Retrospectivos , Neoplasias do Colo/complicações , Neoplasias do Colo/epidemiologia , Neoplasias Colorretais/microbiologia , Cirrose Hepática/complicações , Bacteriemia/complicações , Bacteriemia/epidemiologia , Bacteriemia/microbiologia , Infecções Estreptocócicas/complicações , Infecções Estreptocócicas/epidemiologia , Infecções Estreptocócicas/microbiologia
20.
Front Cell Infect Microbiol ; 13: 1146431, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37234777

RESUMO

Streptococcus pyogenes causes a multitude of local and systemic infections, the most common being pharyngitis in children. Recurrent pharyngeal infections are common and are thought to be due to the re-emergence of intracellular GAS upon completion of antibiotic treatment. The role of colonizing biofilm bacteria in this process is not fully clear. Here, live respiratory epithelial cells were inoculated with broth-grown or biofilm bacteria of different M-types, as well as with isogenic mutants lacking common virulence factors. All M-types tested adhered to and were internalized into epithelial cells. Interestingly, internalization and persistence of planktonic bacteria varied significantly between strains, whereas biofilm bacteria were internalized in similar and higher numbers, and all strains persisted beyond 44 hours, showing a more homogenous phenotype. The M3 protein, but not the M1 or M5 proteins, was required for optimal uptake and persistence of both planktonic and biofilm bacteria inside cells. Moreover, the high expression of capsule and SLO inhibited cellular uptake and capsule expression was required for intracellular survival. Streptolysin S was required for optimal uptake and persistence of M3 planktonic bacteria, whereas SpeB improved intracellular survival of biofilm bacteria. Microscopy of internalized bacteria showed that planktonic bacteria were internalized in lower numbers as individual or small clumps of bacteria in the cytoplasm, whereas GAS biofilm bacteria displayed a pattern of perinuclear localization of bacterial aggregates that affected actin structure. Using inhibitors targeting cellular uptake pathways, we confirmed that planktonic GAS mainly uses a clathrin-mediated uptake pathway that also required actin and dynamin. Clathrin was not involved in biofilm internalization, but internalization required actin rearrangement and PI3 kinase activity, possibly suggesting macropinocytosis. Together these results provide a better understanding of the potential mechanisms of uptake and survival of various phenotypes of GAS bacteria relevant for colonization and recurrent infection.


Assuntos
Infecções Estreptocócicas , Streptococcus pyogenes , Humanos , Streptococcus pyogenes/genética , Sorogrupo , Virulência , Actinas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células Epiteliais/microbiologia , Biofilmes , Fatores de Virulência/metabolismo , Infecções Estreptocócicas/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA