Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 392
Filtrar
1.
Microbiol Spectr ; 12(6): e0362323, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38722155

RESUMEN

The vaginal microbiota plays a pivotal role in reproductive, sexual, and perinatal health and disease. Unlike the well-established connections between diet, metabolism, and the intestinal microbiota, parallel mechanisms influencing the vaginal microbiota and pathogen colonization remain overlooked. In this study, we combine a mouse model of Streptococcus agalactiae strain COH1 [group B Streptococcus (GBS)] vaginal colonization with a mouse model of pubertal-onset obesity to assess diet as a determinant of vaginal microbiota composition and its role in colonization resistance. We leveraged culture-dependent assessment of GBS clearance and culture-independent, sequencing-based reconstruction of the vaginal microbiota in relation to diet, obesity, glucose tolerance, and microbial dynamics across time scales. Our findings demonstrate that excessive body weight gain and glucose intolerance are not associated with vaginal GBS density or timing of clearance. Diets high in fat and low in soluble fiber are associated with vaginal GBS persistence, and changes in vaginal microbiota structure and composition due to diet contribute to GBS clearance patterns in nonpregnant mice. These findings underscore a critical need for studies on diet as a key determinant of vaginal microbiota composition and its relevance to reproductive and perinatal outcomes.IMPORTANCEThis work sheds light on diet as a key determinant influencing the composition of vaginal microbiota and its involvement in group B Streptococcus (GBS) colonization in a mouse model. This study shows that mice fed diets with different nutritional composition display differences in GBS density and timing of clearance in the female reproductive tract. These findings are particularly significant given clear links between GBS and adverse reproductive and neonatal outcomes, advancing our understanding by identifying critical connections between dietary components, factors originating from the intestinal tract, vaginal microbiota, and reproductive outcomes.


Asunto(s)
Dieta , Infecciones Estreptocócicas , Streptococcus agalactiae , Vagina , Vagina/microbiología , Femenino , Animales , Streptococcus agalactiae/crecimiento & desarrollo , Ratones , Infecciones Estreptocócicas/microbiología , Microbiota/fisiología , Obesidad/microbiología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Humanos
2.
BMC Microbiol ; 22(1): 23, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-35026981

RESUMEN

BACKGROUND: Streptococcus agalactiae or Group B Streptococcus (GBS) is an encapsulated gram-positive bacterial pathobiont that commonly colonizes the lower gastrointestinal tract and reproductive tract of human hosts. This bacterium can infect the gravid reproductive tract and cause invasive infections of pregnant patients and neonates. Upon colonizing the reproductive tract, the bacterial cell is presented with numerous nutritional challenges imposed by the host. One strategy employed by the host innate immune system is intoxication of bacterial invaders with certain transition metals such as zinc. METHODOLOGY: Previous work has demonstrated that GBS must employ elegant strategies to circumnavigate zinc stress in order to survive in the vertebrate host. We assessed 30 strains of GBS from diverse isolation sources, capsular serotypes, and sequence types for susceptibility or resistance to zinc intoxication. RESULTS: Invasive strains, such as those isolated from early onset disease manifestations of GBS infection were significantly less susceptible to zinc toxicity than colonizing strains isolated from rectovaginal swabs of pregnant patients. Additionally, capsular type III (cpsIII) strains and the ST-17 and ST-19 strains exhibited the greatest resilience to zinc stress, whereas ST-1 and ST-12 strains as well as those possessing capsular type Ib (cpsIb) were more sensitive to zinc intoxication. Thus, this study demonstrates that the transition metal zinc possesses antimicrobial properties against a wide range of GBS strains, with isolation source, capsular serotype, and sequence type contributing to susceptibility or resistance to zinc stress.


Asunto(s)
Antibacterianos/farmacología , Cloruros/farmacocinética , Serogrupo , Streptococcus agalactiae/efectos de los fármacos , Streptococcus agalactiae/genética , Compuestos de Zinc/farmacocinética , Antibacterianos/metabolismo , Cápsulas Bacterianas/clasificación , Cápsulas Bacterianas/efectos de los fármacos , Cloruros/metabolismo , Femenino , Humanos , Recién Nacido , Pruebas de Sensibilidad Microbiana , Embarazo , Serotipificación , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/tratamiento farmacológico , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/clasificación , Streptococcus agalactiae/crecimiento & desarrollo , Vagina/efectos de los fármacos , Vagina/microbiología , Compuestos de Zinc/metabolismo
3.
Microbiol Spectr ; 9(3): e0105821, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34908468

RESUMEN

The Gram-positive pathogen group B Streptococcus (GBS) is a leading cause of neonatal bacterial infections, preterm birth, and stillbirth. Although maternal GBS vaginal colonization is a risk factor for GBS-associated adverse birth outcomes, mechanisms promoting GBS vaginal persistence are not fully defined. GBS possesses a broadly conserved small molecule, CAMP factor, that is co-hemolytic in the presence of Staphylococcus aureus sphingomyelinase C. While this co-hemolytic reaction is commonly used by clinical laboratories to identify GBS, the contribution of CAMP factor to GBS vaginal persistence is unknown. Using in vitro biofilm, adherence and invasion assays with immortalized human vaginal epithelial VK2 cells, and a mouse model of GBS vaginal colonization, we tested the contribution of CAMP factor using GBS strain COH1 and its isogenic CAMP-deficient mutant (Δcfb). We found no evidence for CAMP factor involvement in GBS biofilm formation, or adherence, invasion, or cytotoxicity toward VK2 cells in the presence or absence of S. aureus. Additionally, there was no difference in vaginal burdens or persistence between COH1 and Δcfb strains in a murine colonization model. In summary, our results using in vitro human cell lines and murine models do not support a critical role for CAMP factor in promoting GBS vaginal colonization. IMPORTANCE Group B Streptococcus (GBS) remains a pervasive pathogen for pregnant women and their newborns. Maternal screening and intrapartum antibiotic prophylaxis to GBS-positive mothers have reduced, but not eliminated GBS neonatal disease, and have not impacted GBS-associated preterm birth or stillbirth. Additionally, this antibiotic exposure is associated with adverse effects on the maternal and neonatal microbiota. Identifying key GBS factors important for maternal vaginal colonization will foster development of more targeted, alternative therapies to antibiotic treatment. Here, we investigate the contribution of a broadly conserved GBS determinant, CAMP factor, to GBS vaginal colonization and find that CAMP factor is unlikely to be a biological target to control maternal GBS colonization.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Proteínas Hemolisinas/metabolismo , Membrana Mucosa/microbiología , Streptococcus agalactiae/metabolismo , Vagina/microbiología , Animales , Adhesión Bacteriana/fisiología , Proteínas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Línea Celular , Células Epiteliales/microbiología , Femenino , Eliminación de Gen , Proteínas Hemolisinas/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Embarazo , Esfingomielina Fosfodiesterasa/metabolismo , Streptococcus agalactiae/genética , Streptococcus agalactiae/crecimiento & desarrollo
4.
PLoS One ; 16(12): e0252973, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34860840

RESUMEN

Camels are vital to food production in the drylands of the Horn of Africa, with milk as their main contribution to food security. A major constraint to camel milk production is mastitis, inflammation of the mammary gland. The condition negatively impacts milk yield and quality as well as household income. A leading cause of mastitis in dairy camels is Streptococcus agalactiae, or group B Streptococcus (GBS), which is also a commensal and pathogen of humans and cattle. It has been suggested that extramammary reservoirs for this pathogen may contribute to the occurrence of mastitis in camels. We explored the molecular epidemiology of GBS in camels using a cross-sectional study design for sample collection and phenotypic, genomic and phylogenetic analysis of isolates. Among 88 adult camels and 93 calves from six herds in Laikipia County, Kenya, GBS was detected in 20% of 50 milk samples, 25% of 152 nasal swabs, 8% of 90 oral swabs and 3% of 90 rectal swabs, but not in vaginal swabs. Per camel herd, two to four sequence types (ST) were identified using Multi Locus Sequence Typing (MLST). More than half of the isolates belonged to ST617 or its single-locus variant, ST1652, with these STs found across all sample types. Capsular serotype VI was detected in 30 of 58 isolates. In three herds, identical STs were detected in milk and swab samples, suggesting that extramammary sources of GBS may contribute to the maintenance and spread of GBS within camel herds. This needs to be considered when developing prevention and control strategies for GBS mastitis. The high nasal carriage rate, low recto-vaginal carriage rate, and high prevalence of serotype VI for GBS in camels are in stark contrast to the distribution of GBS in humans and in cattle and reveal hitherto unknown ecological and molecular features of this bacterial species.


Asunto(s)
Camelus/microbiología , Infecciones Estreptocócicas , Streptococcus agalactiae , Animales , Femenino , Humanos , Kenia/epidemiología , Masculino , Glándulas Mamarias Animales/microbiología , Leche/microbiología , Tipificación de Secuencias Multilocus , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/veterinaria , Streptococcus agalactiae/clasificación , Streptococcus agalactiae/crecimiento & desarrollo
5.
Microbiol Spectr ; 9(1): e0007721, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34378963

RESUMEN

Group B Streptococcus (GBS) causes serious neonatal infection via vertical transmission. The prenatal GBS screening test is performed at the late stage of pregnancy to avoid risks of infection. In this test, enrichment culture is performed, followed by GBS identification. Selective medium is used for the enrichment; however, Enterococcus faecalis, which is a potential contaminant in swab samples, can interfere with the growth of GBS. Such bacterial contamination can lead to false-negative results. Endolysin, a bacteriophage-derived enzyme, degrades peptidoglycan in the bacterial cell wall; it is a promising antimicrobial agent for selectively eliminating specific bacterial genera/species. In this study, we used the recombinant endolysin EG-LYS, which is specific to E. faecalis; the endolysin potentially enriched GBS in the selective culture. First, in the false-negative model (coculture of GBS and E. faecalis, which disabled GBS detection in the subsequent GBS identification test), EG-LYS treatment at 0.1 mg/ml improved GBS detection. Next, we used 548 vaginal swabs to test the efficacy of EG-LYS treatment in improving GBS detection. EG-LYS treatment (0.1 mg/ml) increased the GBS-positive ratio to 17.9%, compared to 15.7% in the control (phosphate-buffered saline [PBS] treatment). In addition, there were an increased number of GBS colonies under EG-LYS treatment in some samples. The results were supported by the microbiota analysis of the enriched cultures. In conclusion, EG-LYS treatment of the enrichment culture potentially improves the accuracy of the prenatal GBS screening test. IMPORTANCE Endolysin is a bacteriophage-derived enzyme that degrades the peptidoglycan in the cell wall of host bacteria; it could be used as an antimicrobial agent for selectively eliminating specific bacterial genera/species. Group B Streptococcus (GBS) causes neonatal infection via vertical transmission; prenatal GBS screening test, in which enrichment culture is followed by bacterial identification, is used to detect the presence of GBS in pregnant women. However, the presence of commensal bacteria such as Enterococcus faecalis in clinical specimens can inhibit GBS growth in the selective enrichment culture, resulting in false-negative result. Here, we demonstrated that the application of originally isolated endolysin in the enrichment culture improved the test accuracy by inhibiting unwanted E. faecalis growth and therefore avoiding false-negative results, not only in experimental settings, but also in tests using vaginal swabs.


Asunto(s)
Endopeptidasas/farmacología , Complicaciones Infecciosas del Embarazo/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/efectos de los fármacos , Bacteriófagos/genética , Bacteriófagos/metabolismo , Medios de Cultivo/metabolismo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Femenino , Humanos , Embarazo , Complicaciones Infecciosas del Embarazo/diagnóstico , Infecciones Estreptocócicas/diagnóstico , Streptococcus agalactiae/crecimiento & desarrollo , Streptococcus agalactiae/aislamiento & purificación , Streptococcus agalactiae/metabolismo , Vagina/microbiología
6.
Fish Shellfish Immunol ; 117: 62-69, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34280520

RESUMEN

This study aimed to verify the effects of dietary supplementation with sodium butyrate and Lippia origanoides, combined and isolated, on the health and zootechnical performance of Nile tilapia juveniles Oreochromis niloticus. A total of 120 fish (5.38 ± 0.65 g) were randomly distributed in 12 experimental units and fed different experimental diets for 30 days, namely: commercial diet without supplementation (Unsupplemented); commercial diet supplemented with 0.5% sodium butyrate (Butyrate); commercial diet supplemented with 0.125% L. origanoides (Lippia) and commercial diet supplemented with a mixture of 0.5% sodium butyrate and 0.125% L. origanoides (Butyrate + Lippia). After preparing the experimental diets there was an increase in the pH of diet Butyrate when compared to the other diets. After 30 days the fish supplemented with Butyrate + Lippia showed reduction significate in the mean corpuscular haemoglobin, concentration of total heterotrophic bacteria in the intestine, and lymphocyte infiltrates in the liver. Besides that, the supplementation with Butyrate + Lippia promoted an increased number of intestinal villi compared to the fish Unsupplemented ones. Additionally, fish fed a diet containing only Lippia presented an increase in the villus perimeter in the posterior region of the gut and in the red blood cell number. Animals supplemented only with sodium butyrate demonstrated increased lactic acid bacterium in the gut and macrosteatosis in the liver, besides decreased melanomacrophages in the spleen. The use of sodium butyrate associated with essential oil had positive effects on the intestinal microbiota, intestinal structure, liver, and spleen integrity, suggesting a greater efficiency of the compounds when used together in the nutrition of Nile tilapia juveniles.


Asunto(s)
Ácido Butírico/farmacología , Cíclidos , Suplementos Dietéticos , Lippia , Aceites Volátiles/farmacología , Animales , Cíclidos/sangre , Cíclidos/crecimiento & desarrollo , Cíclidos/inmunología , Cíclidos/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Pruebas Hematológicas , Hígado/efectos de los fármacos , Hígado/inmunología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Bazo/efectos de los fármacos , Bazo/inmunología , Streptococcus agalactiae/efectos de los fármacos , Streptococcus agalactiae/crecimiento & desarrollo
7.
J Bacteriol ; 203(17): e0023421, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34124943

RESUMEN

High-molecular-mass penicillin-binding proteins (PBPs) are enzymes that catalyze the biosynthesis of bacterial cell wall peptidoglycan. The Gram-positive bacterial pathogen Streptococcus agalactiae (group B streptococcus [GBS]) produces five high-molecular-mass PBPs, namely, PBP1A, PBP1B, PBP2A, PBP2B, and PBP2X. Among these, only PBP2X is essential for cell viability, whereas the other four PBPs are individually dispensable. The biological function of the four nonessential PBPs is poorly characterized in GBS. We deleted the pbp1a, pbp1b, pbp2a, and pbp2b genes individually from a genetically well-characterized serotype V GBS strain and studied the phenotypes of the four isogenic mutant strains. Compared to the wild-type parental strain, (i) none of the pbp isogenic mutant strains had a significant growth defect in Todd-Hewitt broth supplemented with 0.2% yeast extract (THY) rich medium, (ii) isogenic mutant Δpbp1a and Δpbp1b strains had significantly increased susceptibility to penicillin and ampicillin, and (iii) isogenic mutant Δpbp1a and Δpbp2b strains had significantly longer chain lengths. Using saturated transposon mutagenesis and transposon insertion site sequencing, we determined the genes essential for the viability of the wild-type GBS strain and each of the four isogenic pbp deletion mutant strains in THY rich medium. The pbp1a gene is essential for cell viability in the pbp2b deletion background. Reciprocally, pbp2b is essential in the pbp1a deletion background. Moreover, the gene encoding RodA, a peptidoglycan polymerase that works in conjunction with PBP2B, is also essential in the pbp1a deletion background. Together, our results suggest functional overlap between PBP1A and the PBP2B-RodA complex in GBS cell wall peptidoglycan biosynthesis. IMPORTANCE High-molecular-mass penicillin-binding proteins (HMM PBPs) are enzymes required for bacterial cell wall biosynthesis. Bacterial pathogen group B streptococcus (GBS) produces five distinct HMM PBPs. The biological functions of these proteins are not well characterized in GBS. In this study, we performed a comprehensive deletion analysis of genes encoding HMM PBPs in GBS. We found that deleting certain PBP-encoding genes altered bacterial susceptibility to beta-lactam antibiotics, cell morphology, and the essentiality of other enzymes involved in cell wall peptidoglycan synthesis. The results of our study shed new light on the biological functions of PBPs in GBS.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas de Unión a las Penicilinas/genética , Proteínas de Unión a las Penicilinas/metabolismo , Streptococcus agalactiae/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/química , Eliminación de Gen , Mutagénesis , Mutagénesis Insercional , Proteínas de Unión a las Penicilinas/química , Penicilinas/farmacología , Streptococcus agalactiae/efectos de los fármacos , Streptococcus agalactiae/genética , Streptococcus agalactiae/crecimiento & desarrollo
8.
BMC Microbiol ; 21(1): 145, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33985431

RESUMEN

BACKGROUND: Group B streptococci (GBS) are important neonatal bacterial pathogens that can cause severe invasive disease in the newborn. It is thought that in many cases of invasive neonatal GBS disease, the bacteria ascend the vagina into the uterus and infect the amniotic fluid surrounding the fetus. Important constituents of this environment include the polyols or sugar alcohols of which erythritol, sorbitol and mannitol are examples. The aim of our study was to investigate the effect of polyols on GBS grown in media containing these sugar alcohols. RESULTS: GBS incubated in varying concentrations of polyols (erythritol, sorbitol or mannitol) did not display any significant enhancement or inhibition of bacterial growth. However, growth of GBS in the presence of erythritol significantly increased the surface expression of GBS-PGK (a plasminogen binding protein) 1.25 to 1.5-fold depending on the erythritol concentration and significantly enhanced the survival in human blood 3X to 18X depending on the concentration of polyol used. Interestingly, GBS grown in 1% erythritol significantly increased invasion by the bacteria of HeLa cells (epithelial cell line) (150% vs 100%) however, at higher concentrations (2% or 4% of polyol) the number of CFUs was significantly reduced (55-75% vs 100%) suggesting higher concentrations of polyols may inhibit invasion. Erythritol also increased GBS hemolytic activity as well as enhancing biofilm formation 1.4X to 3.3X depending on the concentration of polyol used. CONCLUSIONS: GBS grown in the presence of polyols alters the bacteria's phenotype resulting in changes associated with GBS virulence. This effect was greatest for the polyol erythritol.


Asunto(s)
Eritritol/metabolismo , Manitol/metabolismo , Polímeros/metabolismo , Sorbitol/metabolismo , Streptococcus agalactiae/crecimiento & desarrollo , Células HeLa , Humanos , Fenotipo , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Virulencia
9.
mSphere ; 6(3)2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011683

RESUMEN

Zinc is an essential trace element for normal bacterial physiology but, divergently, can intoxicate bacteria at high concentrations. Here, we define the molecular systems for Zn detoxification in Streptococcus agalactiae, also known as group B streptococcus, and examine the effects of resistance to Zn stress on virulence. We compared the growth of wild-type bacteria and mutants deleted for the Zn exporter, czcD, and the response regulator, sczA, using Zn-stress conditions in vitro Macrophage antibiotic protection assays and a mouse model of disseminated infection were used to assess virulence. Global bacterial transcriptional responses to Zn stress were defined by RNA sequencing and quantitative reverse transcription-PCR. czcD and sczA enabled S. agalactiae to survive Zn stress, with the putative CzcD efflux system activated by SczA. Additional genes activated in response to Zn stress encompassed divalent cation transporters that contribute to regulation of Mn and Fe homeostasis. In vivo, the czcD-sczA Zn management axis supported virulence in the blood, heart, liver, and bladder. Additionally, several genes not previously linked to Zn stress in any bacterium, including, most notably, arcA for arginine deamination, also mediated resistance to Zn stress, representing a novel molecular mechanism of bacterial resistance to metal intoxication. Taken together, these findings show that S. agalactiae responds to Zn stress by sczA regulation of czcD, with additional novel mechanisms of resistance supported by arcA, encoding arginine deaminase. Cellular management of Zn stress in S. agalactiae supports virulence by facilitating bacterial survival in the host during systemic infection.IMPORTANCEStreptococcus agalactiae, also known as group B streptococcus, is an opportunistic pathogen that causes various diseases in humans and animals. This bacterium has genetic systems that enable zinc detoxification in environments of metal stress, but these systems remain largely undefined. Using a combination of genomic, genetic, and cellular assays, we show that this pathogen controls Zn export through CzcD to manage Zn stress and utilizes a system of arginine deamination never previously linked to metal stress responses in bacteria to survive metal intoxication. We show that these systems are crucial for survival of S. agalactiaein vitro during Zn stress and also enhance virulence during systemic infection in mice. These discoveries establish new molecular mechanisms of resistance to metal intoxication in bacteria; we suggest these mechanisms operate in other bacteria as a way to sustain microbial survival under conditions of metal stress, including in host environments.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Metales/farmacología , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/efectos de los fármacos , Streptococcus agalactiae/patogenicidad , Estrés Fisiológico , Zinc/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Humanos , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Streptococcus agalactiae/genética , Streptococcus agalactiae/crecimiento & desarrollo , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Transcripción Genética , Células U937 , Virulencia , Zinc/análisis
10.
Microbiology (Reading) ; 167(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33983874

RESUMEN

Streptococcus pneumoniae, S. pyogenes (Group A Streptococcus; GAS) and S. agalactiae (Group B Streptococcus; GBS) are major aetiological agents of diseases in humans. The cellular membrane, a crucial site in host-pathogen interactions, is poorly characterized in streptococci. Moreover, little is known about whether or how environmental conditions influence their lipid compositions. Using normal phase liquid chromatography coupled with electrospray ionization MS, we characterized the phospholipids and glycolipids of S. pneumoniae, GAS and GBS in routine undefined laboratory medium, streptococcal defined medium and, in order to mimic the host environment, defined medium supplemented with human serum. In human serum-supplemented medium, all three streptococcal species synthesize phosphatidylcholine (PC), a zwitterionic phospholipid commonly found in eukaryotes but relatively rare in bacteria. We previously reported that S. pneumoniae utilizes the glycerophosphocholine (GPC) biosynthetic pathway to synthesize PC. Through substrate tracing experiments, we confirm that GAS and GBS scavenge lysoPC, a major metabolite in human serum, thereby using an abbreviated GPC pathway for PC biosynthesis. Furthermore, we found that plasmanyl-PC is uniquely present in the GBS membrane during growth with human serum, suggesting GBS possesses unusual membrane biochemical or biophysical properties. In summary, we report cellular lipid remodelling by the major pathogenic streptococci in response to metabolites present in human serum.


Asunto(s)
Fosfolípidos/metabolismo , Suero/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/metabolismo , Streptococcus pneumoniae/metabolismo , Streptococcus pyogenes/metabolismo , Membrana Celular/química , Membrana Celular/genética , Medios de Cultivo/metabolismo , Humanos , Fosfolípidos/química , Infecciones Estreptocócicas/sangre , Streptococcus agalactiae/química , Streptococcus agalactiae/crecimiento & desarrollo , Streptococcus pneumoniae/química , Streptococcus pneumoniae/crecimiento & desarrollo , Streptococcus pyogenes/química , Streptococcus pyogenes/crecimiento & desarrollo
11.
Gynecol Endocrinol ; 36(sup1): 48-52, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33305673

RESUMEN

OBJECTIVE: The objective of our study was to explore the possible graphene impact on microorganism growth as well as on laboratory animal overall condition. Materials and technique: The experiments applied samples of graphene three concentrations and two 15 × 15 mm quartz glasses one of which carrying deposited graphene lattice. We have also used 5% blood agar, thioglycollate broth, bacterial suspensions of standard turbidity containing pure clinical isolate of microorganisms. Three white male 6-month-old laboratory rats were used to estimate the graphene impact on the overall animal condition. RESULTS: Graphene did not contain any microorganisms, does not destroy erythrocytes placed within the artificial nutritional medium, graphene lattice did not add any properties to the quartz glasses which could allow the Proteus spread all over its surface. It was also established that graphene did not show any native antibacterial impact. No significant reaction was noticed in animals after graphene administration to laboratory rats neither at the injection spot nor at the overall level. CONCLUSION: Our data confirm the applicability of graphene both in scientific and practical biomedical purposes.


Asunto(s)
Grafito/toxicidad , Prótesis e Implantes/efectos adversos , Animales , Escherichia coli/crecimiento & desarrollo , Grafito/química , Ensayo de Materiales , Medicina/métodos , Pruebas de Sensibilidad Microbiana , Ratas , Staphylococcus aureus/crecimiento & desarrollo , Streptococcus agalactiae/crecimiento & desarrollo , Pruebas de Toxicidad
12.
mBio ; 11(6)2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33173000

RESUMEN

Nutritional immunity is an elegant host mechanism used to starve invading pathogens of necessary nutrient metals. Calprotectin, a metal-binding protein, is produced abundantly by neutrophils and is found in high concentrations within inflammatory sites during infection. Group B Streptococcus (GBS) colonizes the gastrointestinal and female reproductive tracts and is commonly associated with severe invasive infections in newborns such as pneumonia, sepsis, and meningitis. Although GBS infections induce robust neutrophil recruitment and inflammation, the dynamics of GBS and calprotectin interactions remain unknown. Here, we demonstrate that disease and colonizing isolate strains exhibit susceptibility to metal starvation by calprotectin. We constructed a mariner transposon (Krmit) mutant library in GBS and identified 258 genes that contribute to surviving calprotectin stress. Nearly 20% of all underrepresented mutants following treatment with calprotectin are predicted metal transporters, including known zinc systems. As calprotectin binds zinc with picomolar affinity, we investigated the contribution of GBS zinc uptake to overcoming calprotectin-imposed starvation. Quantitative reverse transcriptase PCR (qRT-PCR) revealed a significant upregulation of genes encoding zinc-binding proteins, adcA, adcAII, and lmb, following calprotectin exposure, while growth in calprotectin revealed a significant defect for a global zinc acquisition mutant (ΔadcAΔadcAIIΔlmb) compared to growth of the GBS wild-type (WT) strain. Furthermore, mice challenged with the ΔadcAΔadcAIIΔlmb mutant exhibited decreased mortality and significantly reduced bacterial burden in the brain compared to mice infected with WT GBS; this difference was abrogated in calprotectin knockout mice. Collectively, these data suggest that GBS zinc transport machinery is important for combatting zinc chelation by calprotectin and establishing invasive disease.IMPORTANCE Group B Streptococcus (GBS) asymptomatically colonizes the female reproductive tract but is a common causative agent of meningitis. GBS meningitis is characterized by extensive infiltration of neutrophils carrying high concentrations of calprotectin, a metal chelator. To persist within inflammatory sites and cause invasive disease, GBS must circumvent host starvation attempts. Here, we identified global requirements for GBS survival during calprotectin challenge, including known and putative systems involved in metal ion transport. We characterized the role of zinc import in tolerating calprotectin stress in vitro and in a mouse model of infection. We observed that a global zinc uptake mutant was less virulent than the parental GBS strain and found calprotectin knockout mice to be equally susceptible to infection by wild-type (WT) and mutant strains. These findings suggest that calprotectin production at the site of infection results in a zinc-limited environment and reveals the importance of GBS metal homeostasis to invasive disease.


Asunto(s)
Complejo de Antígeno L1 de Leucocito/metabolismo , Infecciones Estreptocócicas/metabolismo , Streptococcus agalactiae/metabolismo , Zinc/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Femenino , Humanos , Complejo de Antígeno L1 de Leucocito/genética , Meningitis Bacterianas/genética , Meningitis Bacterianas/metabolismo , Meningitis Bacterianas/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/genética , Streptococcus agalactiae/crecimiento & desarrollo , Streptococcus agalactiae/patogenicidad , Virulencia
13.
Res Microbiol ; 171(8): 311-318, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32896574

RESUMEN

Bacterial biofilms are an important virulence factor with a vital role in evasion from the host immune system, colonization and infection. The aim of the present study was to evaluate in vitro the effects of three environmental factors (H+, glucose and human plasma) in biofilm formation, by carrier and invasive Streptococcus agalactiae strains of ST17 and ST19 sequence types, including DNase producers and non-producers. Bacteria ability to assemble biofilms was classified based on crystal violet assay. Biofilm formation was also monitored by scanning electron microscopy. Depending on the growth medium used, each bacterial isolate could fit in different biofilm production categories. Our data showed that optimal conditions for S. agalactiae biofilm assembly were reached after 48 h incubation at pH 7.6 in the presence of glucose and inactivated human plasma. In the presence of inactivated human plasma, the biofilm biomass of ST19 strains experienced a higher increase than ST17 strains. The composition of the extracellular polymeric matrix of the three strongest biofilm producers (all from ST17) was accessed by enzymatic digestion of mature biofilms and proteins were shown to be the predominant component. The detailed identification of the extracellular protein components should contribute to the development of new therapeutic strategies to fight S. agalactiae infections.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Streptococcus agalactiae/crecimiento & desarrollo , Medios de Cultivo/metabolismo , Matriz Extracelular de Sustancias Poliméricas/química , Glucosa , Humanos , Concentración de Iones de Hidrógeno , Microscopía Electrónica de Rastreo , Plasma , Streptococcus agalactiae/ultraestructura , Factores de Virulencia/metabolismo
14.
Sci Rep ; 10(1): 15208, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32938989

RESUMEN

Treatment of bacterial infections is a great challenge of our era due to the various resistance mechanisms against antibiotics. Antimicrobial peptides are considered to be potential novel compound as antibiotic treatment. However, some bacteria, especially many human pathogens, are inherently resistant to these compounds, due to the expression of BceAB-type ABC transporters. This rather new transporter family is not very well studied. Here, we report the first full characterization of the nucleotide binding domain of a BceAB type transporter from Streptococcus agalactiae, namely SaNsrF of the transporter SaNsrFP, which confers resistance against nisin and gallidermin. We determined the NTP hydrolysis kinetics and used molecular modeling and simulations in combination with small angle X-ray scattering to obtain structural models of the SaNsrF monomer and dimer. The fact that the SaNsrFH202A variant displayed no ATPase activity was rationalized in terms of changes of the structural dynamics of the dimeric interface. Kinetic data show a clear preference for ATP as a substrate, and the prediction of binding modes allowed us to explain this selectivity over other NTPs.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Farmacorresistencia Bacteriana Múltiple , Streptococcus agalactiae/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Bacteriocinas/farmacología , Sitios de Unión , Clonación Molecular , Regulación Bacteriana de la Expresión Génica , Hidrólisis , Modelos Moleculares , Simulación del Acoplamiento Molecular , Nisina/farmacología , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Dispersión del Ángulo Pequeño , Streptococcus agalactiae/química , Streptococcus agalactiae/genética , Streptococcus agalactiae/crecimiento & desarrollo , Difracción de Rayos X
15.
Anim Reprod Sci ; 222: 106602, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32980651

RESUMEN

Endometritis represents the main cause of reproductive failure in dromedary camels. In dromedary camels, associations between endometritis-causing pathogen-species, disease severity, and systemic changes in the immune system have not been evaluated. In the current study, there was use of flow cytometry and immunofluorescence of membrane proteins for the evaluation of leukocyte subsets and the cellular phenotype in blood of camels with clinical endometritis and evaluations of associations with disease severity and endometritis-causing pathogens. Animals with endometritis had markedly larger numbers of total leukocytes and neutrophils. Although total lymphocyte and monocyte counts did not differ between camels with and without clinical endometritis, there were lesser numbers of total and effector CD4-positive T cells in camels with endometritis. Among monocytes, number of camel inflammatory monocytes (Mo-II) was markedly greater, whereas Mo-III numbers were less in the blood of camels with clinical endometritis. Number of inflammatory monocytes was also indicative of endometritis severity grade. Among camels with clinical endometritis, E. coli- and S. aureus-infected animals had similar endometritis grades and comparable phenotype and composition patterns of leukocytes. Neutrophils and monocytes of camels with clinical endometritis had fewer cell adhesion molecules (i.e., CD11a and CD18). Collectively, the results from the current study allowed for identification of associations between endometritis severity grade and larger numbers of inflammatory monocytes. The results also indicate there is no association between endometritis pathogen-species and changes in phenotype or composition of blood leukocytes.


Asunto(s)
Camelus/sangre , Endometritis/veterinaria , Leucocitos/clasificación , Actinomycetaceae/aislamiento & purificación , Animales , Endometritis/sangre , Endometritis/patología , Endometrio/microbiología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/aislamiento & purificación , Femenino , Citometría de Flujo/veterinaria , Leucocitos/citología , Linfocitos/clasificación , Linfocitos/citología , Proteus/aislamiento & purificación , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/aislamiento & purificación , Streptococcus agalactiae/crecimiento & desarrollo , Streptococcus agalactiae/aislamiento & purificación
16.
Infect Immun ; 88(10)2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32747604

RESUMEN

Streptococcus agalactiae (group B streptococcus, or GBS) is a common cause of bacteremia and sepsis in newborns, pregnant women, and immunocompromised patients. The molecular mechanisms used by GBS to survive and proliferate in blood are not well understood. Here, using a highly virulent GBS strain and transposon-directed insertion site sequencing (TraDIS), we performed genome-wide screens to discover novel GBS genes required for bacterial survival in human whole blood and plasma. The screen identified 85 and 41 genes that are required for GBS growth in whole blood and plasma, respectively. A common set of 29 genes was required in both whole blood and plasma. Targeted gene deletion confirmed that (i) genes encoding methionine transporter (metP) and manganese transporter (mtsA) are crucial for GBS survival in whole blood and plasma, (ii) gene W903_1820, encoding a small multidrug export family protein, contributes significantly to GBS survival in whole blood, (iii) the shikimate pathway gene aroA is essential for GBS growth in whole blood and plasma, and (iv) deletion of srr1, encoding a fibrinogen-binding adhesin, increases GBS survival in whole blood. Our findings provide new insight into the GBS-host interactions in human blood.


Asunto(s)
Bacteriemia/microbiología , Genes Bacterianos , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/genética , Proteínas Bacterianas/genética , Aptitud Genética , Genoma Bacteriano/genética , Humanos , Viabilidad Microbiana/genética , Mutagénesis Insercional , Mutación , Streptococcus agalactiae/crecimiento & desarrollo , Streptococcus agalactiae/patogenicidad , Virulencia/genética
17.
Biochem J ; 477(12): 2401-2419, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32519742

RESUMEN

Toxin-antitoxin systems play key roles in bacterial adaptation, including protection from antibiotic assault and infection by bacteriophages. The type IV toxin-antitoxin system AbiE encodes a DUF1814 nucleotidyltransferase-like toxin, and a two-domain antitoxin. In Streptococcus agalactiae, the antitoxin AbiEi negatively autoregulates abiE expression through positively co-operative binding to inverted repeats within the promoter. The human pathogen Mycobacterium tuberculosis encodes four DUF1814 putative toxins, two of which have antitoxins homologous to AbiEi. One such M. tuberculosis antitoxin, named Rv2827c, is required for growth and whilst the structure has previously been solved, the mode of regulation is unknown. To complete the gaps in our understanding, we first solved the structure of S. agalactiae AbiEi to 1.83 Šresolution for comparison with M. tuberculosis Rv2827c. AbiEi contains an N-terminal DNA binding domain and C-terminal antitoxicity domain, with bilateral faces of opposing charge. The overall AbiEi fold is similar to Rv2827c, though smaller, and with a 65° difference in C-terminal domain orientation. We further demonstrate that, like AbiEi, Rv2827c can autoregulate toxin-antitoxin operon expression. In contrast with AbiEi, the Prv2827c promoter contains two sets of inverted repeats, which bind Rv2827c with differing affinities depending on the sequence consensus. Surprisingly, Rv2827c bound with negative co-operativity to the full Prv2827c promoter, demonstrating an unexpectedly complex form of transcriptional regulation.


Asunto(s)
Antitoxinas/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Homeostasis , Secuencias Invertidas Repetidas , Sistemas Toxina-Antitoxina/genética , Antitoxinas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Moleculares , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/metabolismo , Operón , Regiones Promotoras Genéticas , Conformación Proteica , Streptococcus agalactiae/genética , Streptococcus agalactiae/crecimiento & desarrollo , Streptococcus agalactiae/metabolismo
18.
Chemistry ; 26(44): 9923-9928, 2020 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-32084298

RESUMEN

Ellagic acid derivatives possess antimicrobial and antibiofilm properties across a wide-range of microbial pathogens. Due to their poor solubility and ambident reactivity it is challenging to synthesize, purify, and characterize the activity of ellagic acid glycosides. In this study, we have synthesized three ellagic acid glycoconjugates and evaluated their antimicrobial and antibiofilm activity in Streptococcus agalactiae (Group B Streptococcus, GBS). Their significant impacts on biofilm formation were examined via SEM to reveal early-stage inhibition of cellular adhesion. Additionally, the synthetic glycosides were evaluated against five of the six ESKAPE pathogens and two fungal pathogens. These studies reveal that the ellagic acid glycosides possess inhibitory effects on the growth of gram-negative pathogens.


Asunto(s)
Antibacterianos , Biopelículas/efectos de los fármacos , Ácido Elágico/síntesis química , Ácido Elágico/farmacología , Glicósidos/síntesis química , Glicósidos/farmacología , Streptococcus agalactiae/efectos de los fármacos , Streptococcus agalactiae/ultraestructura , Antibacterianos/síntesis química , Antibacterianos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Pruebas de Sensibilidad Microbiana , Microscopía Electrónica de Rastreo , Streptococcus agalactiae/crecimiento & desarrollo
19.
Carbohydr Res ; 488: 107895, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31901817

RESUMEN

Human milk oligosaccharides (HMOs) are a structurally diverse class of carbohydrates that possess strong antibacterial activity against Streptococcus agalactiae (Group B Strep, GBS). This work highlights the design, synthesis, and retained biological activity of several HMO bioorthogonal probes within GBS, a first in class advance. The use of such probes will assist in identifying HMO-protein interactions within GBS and may be broadly applicable in researching HMO cellular targets within a variety of biological systems. Finally, this strategy is highly amenable to other oligosaccharide scaffolds, requiring minimal synthetic transformations and chemical perturbation.


Asunto(s)
Antibacterianos/síntesis química , Leche Humana/química , Oligosacáridos/química , Bibliotecas de Moléculas Pequeñas/síntesis química , Streptococcus agalactiae/crecimiento & desarrollo , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Diseño de Fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Streptococcus agalactiae/efectos de los fármacos , Streptococcus agalactiae/metabolismo
20.
Biofouling ; 35(8): 938-944, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31646898

RESUMEN

Streptococcus agalactiae (group B Streptococcus, GBS) is a major pathogen in humans and animals. Pili and biofilm may be important virulence factors in this bacterial species. Here, biofilm production and the distribution of pilus variants among 134 GBS isolates from human and animal sources were evaluated. Biofilm production was significantly enhanced in 1% glucose-supplemented medium (p < 0.05). Using this medium, most GBS strains were strong biofilm producers. Biomass was mainly composed of proteins, followed by extracellular DNA, while polysaccharides represented a minor portion. All GBS strains presented at least one pilus variant. PI-2a was the most common among human GBS while PI-2b was the most common among animal isolates. Human GBS harboring PI-2b and animal GBS harboring PI-2a presented significantly reduced biofilm production (p = 0.0033). In conclusion, strong biofilm production seems to be a common characteristic in GBS, and association of the clinical source with the pilus variant may be crucial for this.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Fimbrias Bacterianas/genética , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/aislamiento & purificación , Animales , Proteínas Bacterianas/genética , ADN Bacteriano , Variación Genética , Humanos , Streptococcus agalactiae/genética , Streptococcus agalactiae/crecimiento & desarrollo , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA