Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
J Med Microbiol ; 71(2)2022 Feb.
Article in English | MEDLINE | ID: mdl-35113780

ABSTRACT

Although anaerobic bacteria exist in abundance in cystic fibrosis (CF) airways, their role in disease progression is poorly understood. We hypothesized that the presence and relative abundance of the most prevalent, live, anaerobic bacteria in sputum of adults with CF were associated with adverse clinical outcomes. This is the first study to prospectively investigate viable anaerobic bacteria present in the sputum microbiota and their relationship with long-term outcomes in adults with CF. We performed 16S rRNA analysis using a viability quantitative PCR technique on sputum samples obtained from a prospective cohort of 70 adults with CF and collected clinical data over an 8 year follow-up period. We examined the associations of the ten most abundant obligate anaerobic bacteria present in the sputum with annual rate of FEV1 change. The presence of Porphyromonas pasteri and Prevotella nanceiensis were associated with a greater annual rate of FEV1 change; -52.3 ml yr-1 (95 % CI-87.7;-16.9), -67.9 ml yr-1 (95 % CI-115.6;-20.1), respectively. Similarly, the relative abundance of these live organisms were associated with a greater annual rate of FEV1 decline of -3.7 ml yr-1 (95 % CI: -6.1 to -1.3, P=0.003) and -5.3 ml yr-1 (95 % CI: -8.7 to -1.9, P=0.002) for each log2 increment of abundance, respectively. The presence and relative abundance of certain anaerobes in the sputum of adults with CF are associated with a greater rate of long-term lung function decline. The pathogenicity of anaerobic bacteria in the CF airways should be confirmed with further longitudinal prospective studies with a larger cohort of participants.


Subject(s)
Cystic Fibrosis , Microbiota , Porphyromonas , Prevotella , Adult , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Humans , Lung/physiopathology , Porphyromonas/isolation & purification , Porphyromonas/pathogenicity , Prevotella/isolation & purification , Prevotella/pathogenicity , Prospective Studies , RNA, Ribosomal, 16S/genetics , Sputum/microbiology
2.
Sci Rep ; 11(1): 18486, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34531508

ABSTRACT

The prevalence of coronary artery disease (CAD) in Tibetan Highlanders is lower than that in plain-living individuals, but the mechanism still unclear. Gut microbiota (GM) disorder is considered one of the potential factors involved in the pathogenesis of CAD, but the GM characteristics of Tibetan Highlanders suffering from CAD are unknown. We sequenced the V3-V4 region of the 16S ribosomal RNA of gut bacteria from fecal samples from Tibetan and Han CAD patients and healthy individuals inhabiting the Qinghai-Tibet Plateau, as well as from Han CAD patients and healthy individuals living at sea level, and we analyzed the GM characteristics of these subjects by bioinformatics analysis. The results showed that Tibetan Highlanders suffering from CAD had higher GM α-diversity, with differently distributed cluster compared with healthy Tibetan Highlanders and Han CAD patients living at high and low altitudes. Genera Catenibacterium, Clostridium_sensu_stricto, Holdemanella, and Ruminococcus 2 were enriched in Tibetan Highlanders suffering from CAD compared with healthy Tibetan Highlanders and Han CAD patients living at high- and low-altitudes. Prevotella was enriched in Tibetan Highlanders suffering from CAD compared with Han CAD patients living at high- and low-altitudes. Moreover, Catenibacterium was positively correlated with Prevotella. Additionally, Catenibacterium, Holdemanella, and Prevotella were positively correlated with fermented dairy product, carbohydrate and fiber intake by the subjects, while Clostridium_sensu_stricto was negatively correlated with protein intake by the subjects. In conclusion, our study indicated that Tibetan Highlanders suffering from CAD showed distinct GM, which was linked to their unique dietary characteristics and might associated with CAD.


Subject(s)
Altitude , Coronary Artery Disease/microbiology , Diet , Gastrointestinal Microbiome , Aged , Clostridium/isolation & purification , Clostridium/pathogenicity , Coronary Artery Disease/metabolism , Dietary Fiber/metabolism , Dietary Proteins/metabolism , Female , Humans , Male , Middle Aged , Prevotella/isolation & purification , Prevotella/pathogenicity , Ruminococcus/isolation & purification , Ruminococcus/pathogenicity , Tibet
3.
J Virol ; 95(18): e0047921, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34232744

ABSTRACT

HIV-1 elite controllers (EC) are a rare group among HIV-1-infected individuals who can naturally control viral replication for a prolonged period. Due to their heterogeneous nature, no universal mechanism could be attributed to the EC status; instead, several host and viral factors have been discussed as playing a role. In this study, we investigated the fecal metabolome and microbiome in a Swedish cohort of EC (n = 14), treatment-naive viremic progressors (VP; n = 16), and HIV-negative individuals (HC; n = 12). Fecal untargeted metabolomics was performed by four ultra-high-performance liquid chromatography tandem mass spectrometry (UHPLC-MS/MS). Molecular docking and biochemical microscale thermophoresis (MST) were used to describe the peptide-metabolite interactions. Single-cycle infectivity assays were performed in TZM-Bl cell lines using CCR5- and CXCR4-tropic HIV-1 strains. The microbiome analysis was performed using 16S rRNA sequencing. Th effects of metabolites on bacterial species viability were determined using several clinical isolates. We observed an enrichment of dipeptides in EC compared to VP and HC (adjusted P < 0.05). In silico analysis by molecular docking, in vitro biochemical assays, and ex vivo infection assays identified anti-HIV-1 properties for two dipeptides (WG and VQ) that could bind to the HIV-1 gp120, of which WG was more potent. The microbiome analysis identified enrichment of the genus Prevotella in EC, and these dipeptides supported bacterial growth of the genus Prevotella in vitro. The enrichments of the dipeptides and higher abundance of Prevotella have a distinct mechanism of elite control status in HIV-1 infection that influences host metabolism. IMPORTANCE HIV-1 elite controllers (EC) are a rare group among HIV-1-infected individuals who can naturally control viral replication for a prolonged period. Due to their heterogeneous nature, no universal mechanism could be attributed to the EC status; instead, several host and viral factors have been discussed as playing a role. In this study, we investigated the fecal metabolome and microbiome in a Swedish cohort of EC, treatment-naive viremic progressors (VP), and HIV-negative individuals (HC). We observed an enrichment of dipeptides in EC compared to the other two study groups. In silico and in vitro analyses identified anti-HIV-1 properties for two dipeptides that could bind to the HIV-1 gp120 and act as an HIV-1 antagonist. Furthermore, these dipeptides supported bacterial growth of the genus Prevotella in vitro that was enriched in EC, which influences host metabolism. Thus, increased levels of both dipeptides and Prevotella could provide beneficial effects for EC.


Subject(s)
Bacteroidaceae Infections/microbiology , Dipeptides/pharmacology , Feces/microbiology , HIV Infections/prevention & control , HIV-1/physiology , Metabolome , Prevotella/pathogenicity , Adult , Bacteroidaceae Infections/drug therapy , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Cohort Studies , Feces/chemistry , Female , Gene Expression Profiling , HIV Infections/virology , HIV-1/drug effects , Humans , Male , Middle Aged , Molecular Docking Simulation , Phenotype , Virus Replication
4.
Pathobiology ; 88(2): 156-169, 2021.
Article in English | MEDLINE | ID: mdl-33588422

ABSTRACT

Identifying a microbiome pattern in gastric cancer (GC) is hugely debatable due to the variation resulting from the diversity of the studied populations, clinical scenarios, and metagenomic approach. H. pylori remains the main microorganism impacting gastric carcinogenesis and seems necessary for the initial steps of the process. Nevertheless, an additional non-H. pylori microbiome pattern is also described, mainly at the final steps of the carcinogenesis. Unfortunately, most of the presented results are not reproducible, and there are no consensual candidates to share the H. pylori protagonists. Limitations to reach a consistent interpretation of metagenomic data include contamination along every step of the process, which might cause relevant misinterpretations. In addition, the functional consequences of an altered microbiome might be addressed. Aiming to minimize methodological bias and limitations due to small sample size and the lack of standardization of bioinformatics assessment and interpretation, we carried out a comprehensive analysis of the publicly available metagenomic data from various conditions relevant to gastric carcinogenesis. Mainly, instead of just analyzing the results of each available publication, a new approach was launched, allowing the comprehensive analysis of the total sample amount, aiming to produce a reliable interpretation due to using a significant number of samples, from different origins, in a standard protocol. Among the main results, Helicobacter and Prevotella figured in the "top 6" genera of every group. Helicobacter was the first one in chronic gastritis (CG), gastric cancer (GC), and adjacent (ADJ) groups, while Prevotella was the leader among healthy control (HC) samples. Groups of bacteria are differently abundant in each clinical situation, and bacterial metabolic pathways also diverge along the carcinogenesis cascade. This information may support future microbiome interventions aiming to face the carcinogenesis process and/or reduce GC risk.


Subject(s)
Gastrointestinal Microbiome/genetics , Stomach Neoplasms/microbiology , Computational Biology , Gastric Mucosa/microbiology , Gastrointestinal Microbiome/physiology , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Humans , Metabolic Networks and Pathways , Metagenome , Prevotella/genetics , Prevotella/pathogenicity
5.
Genomics ; 113(1 Pt 2): 664-676, 2021 01.
Article in English | MEDLINE | ID: mdl-33010388

ABSTRACT

Although the prevalence of inflammatory bowel disease (IBD) has been increasing worldwide, the etiology remains elusive. Investigating oral microbiota dysbiosis is essential to understanding IBD pathogenesis. Our study evaluated variations in salivary microbiota and identified potential associations with IBD. The saliva microbiota of 22 IBD patients and 8 healthy controls (HCs) was determined using 16S ribosomal RNA (rRNA) gene sequencing and analyzed using QIIME2. A distinct saliva microbiota dysbiosis in IBD, characterized by alterations in microbiota biodiversity and composition, was identified. Saccharibacteria (TM7), Absconditabacteria (SR1), Leptotrichia, Prevotella, Bulleidia, and Atopobium, some of which are oral biofilm-forming bacteria, were significantly increased. Moreover, levels of inflammatory cytokines associated with IBD were elevated and positively correlated with TM7 and SR1. Functional variations include down-regulation of genetic information processing, while up-regulation of carbohydrate metabolism and protein processing in the endoplasmic reticulum in IBD. Our data implicate salivary microbiota dysbiosis involving in IBD pathogenesis.


Subject(s)
Dysbiosis/microbiology , Inflammatory Bowel Diseases/microbiology , Metagenome , Mouth/microbiology , Adult , Dysbiosis/complications , Dysbiosis/epidemiology , Female , Gastrointestinal Microbiome , Humans , Inflammatory Bowel Diseases/complications , Leptotrichia/genetics , Leptotrichia/pathogenicity , Male , Prevotella/genetics , Prevotella/pathogenicity
6.
PLoS Comput Biol ; 16(12): e1008473, 2020 12.
Article in English | MEDLINE | ID: mdl-33315858

ABSTRACT

Changes in the composition of the microbiome over time are associated with myriad human illnesses. Unfortunately, the lack of analytic techniques has hindered researchers' ability to quantify the association between longitudinal microbial composition and time-to-event outcomes. Prior methodological work developed the joint model for longitudinal and time-to-event data to incorporate time-dependent biomarker covariates into the hazard regression approach to disease outcomes. The original implementation of this joint modeling approach employed a linear mixed effects model to represent the time-dependent covariates. However, when the distribution of the time-dependent covariate is non-Gaussian, as is the case with microbial abundances, researchers require different statistical methodology. We present a joint modeling framework that uses a negative binomial mixed effects model to determine longitudinal taxon abundances. We incorporate these modeled microbial abundances into a hazard function with a parameterization that not only accounts for the proportional nature of microbiome data, but also generates biologically interpretable results. Herein we demonstrate the performance improvements of our approach over existing alternatives via simulation as well as a previously published longitudinal dataset studying the microbiome during pregnancy. The results demonstrate that our joint modeling framework for longitudinal microbiome count data provides a powerful methodology to uncover associations between changes in microbial abundances over time and the onset of disease. This method offers the potential to equip researchers with a deeper understanding of the associations between longitudinal microbial composition changes and disease outcomes. This new approach could potentially lead to new diagnostic biomarkers or inform clinical interventions to help prevent or treat disease.


Subject(s)
Disease , Microbiota , Models, Statistical , Models, Theoretical , Biomarkers/analysis , Female , Humans , Longitudinal Studies , Pre-Eclampsia/microbiology , Pregnancy , Prevotella/isolation & purification , Prevotella/pathogenicity , Proportional Hazards Models , Software
7.
PLoS One ; 15(11): e0242091, 2020.
Article in English | MEDLINE | ID: mdl-33166362

ABSTRACT

BACKGROUND: Perioperative oral management has been reported to be effective for preventing postoperative infectious complications. In addition, severe periodontal disease was identified as the significant risk factor for complications after gastrointestinal surgery. We investigated the bacteriological association between the periodontal pocket, stomach mucosa and drainage fluid to determine whether oral bacteria directly cause intra-abdominal infection after gastrectomy. METHODS: Patients who were scheduled to undergo surgery for gastric cancer were prospectively enrolled. We evaluated the similarity of bacterial strains in periodontal pocket, stomach mucosa and fluid from drainage tube. Gingival crevicular fluid and dental plaque were collected from the periodontal pocket and cultured to detect bacteria. Specimens from the resected stomach were collected and used for bacterial culturing. Drainage fluid from the abdominal cavity was also cultured. RESULTS: All of 52 patients were enrolled. In the periodontal pocket, α-Streptococcus spp., Neisseria sp., and Prevotella sp. were mainly detected. Bacterial cultures in the stomach mucosa were positive in 26 cases. In 20 cases (76.9%), the detected strains were the same as those in the periodontal pocket. Six patients had the postoperative intra-abdominal infection after gastrectomy, and the same bacterial strains was detected in both of drainage fluid and periodontal pocket in two patients with severe periodontal disease. CONCLUSIONS: We found the bacteriological association that same strain detected in periodontal pocket, stomach and in intra-abdominal drainage fluid after gastrectomy in patients with periodontal disease.


Subject(s)
Abdominal Abscess/epidemiology , Gastrectomy/adverse effects , Periodontitis/epidemiology , Postoperative Complications/epidemiology , Abdominal Abscess/microbiology , Aged , Female , Gastric Mucosa/microbiology , Humans , Male , Mouth Mucosa/microbiology , Neisseria/pathogenicity , Periodontitis/microbiology , Postoperative Complications/microbiology , Prevotella/pathogenicity , Streptococcus/pathogenicity
8.
J Infect Dis ; 222(12): 2082-2092, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32515473

ABSTRACT

BACKGROUND: Prevotella species are commonly isolated from the reproductive tract of women with obstetric/gynecologic health complications. However, contributions of this genus to changes in local microenvironment are not well characterized. Our objective was to evaluate species-specific effects of Prevotella on the human endometrial epithelium. METHODS: Thirteen Prevotella strains, originally isolated from the human oral cavity, amniotic fluid, endometrium, or vagina (including women with bacterial vaginosis), were obtained from BEI and ATCC resources. Bacteria were evaluated in silico and in vitro using human endometrial epithelial cells (EEC) grown as monolayers or a 3-dimensional (3D) model. RESULTS: Genomic characterization illustrated metabolic and phylogenetic diversity of Prevotella genus. Among tested species, P. disiens exhibited cytotoxicity. Scanning electron microscopy analysis of the 3D EEC model revealed species-specific colonization patterns and alterations of ultracellular structures. Infection with sialidase-producing P. timonensis resulted in elongated microvilli, and increased MUC3 and MUC4 expression. Infections with Prevotella species, including P. bivia, did not result in significant proinflammatory activation of EEC. CONCLUSIONS: Collectively, findings indicate that Prevotella species are metabolically diverse and overall not cytotoxic or overtly inflammatory in EEC; however, these bacteria can form biofilms, alter barrier properties of the endometrial epithelium, and ultimately impact colonization of secondary colonizers.


Subject(s)
Epithelial Cells/microbiology , Immunity, Innate , Prevotella/genetics , Prevotella/pathogenicity , Cell Line, Tumor , Endometrium/cytology , Epithelial Cells/immunology , Female , Humans , Microscopy, Electron, Scanning , Mucins/genetics , Prevotella/immunology , Species Specificity
9.
Sci Rep ; 10(1): 6365, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286402

ABSTRACT

To construct a saliva-based caries risk assessment model, saliva samples from 176 severe early childhood caries (S-ECC) children and 178 healthy (H) children were screened by real-time PCR-based quantification of the selected species, including Streptococcus mutans, Prevotella pallens, Prevotella denticola and Lactobacillus fermentum. Host factors including caries status, dmft indices, age, gender, and geographic origin were assessed in their influence on abundance of the targeted species, which revealed host caries status as the dominant factor, followed by dmft indices (both P < 0.01). Moreover, levels of S. mutans and P. denticola in the S-ECC group were significantly higher than those in the healthy group (P < 0.001 for S. mutans and P < 0.01 for P. denticola). Interestingly, the co-occurrence network of these targeted species in the S-ECC group differed from that from the healthy group. Finally, based on the combined change pattern of S. mutans and P. pallens, we constructed an S-ECC diagnosis model with an accuracy of 72%. This saliva-based caries diagnosis model is of potential value for circumstances where sampling dental plague is difficult.


Subject(s)
Dental Caries/genetics , Dental Caries/microbiology , Microbiota/genetics , Saliva/microbiology , Child , Child, Preschool , Dental Caries/epidemiology , Dental Caries/pathology , Female , Humans , Limosilactobacillus fermentum/genetics , Limosilactobacillus fermentum/pathogenicity , Male , Prevotella/genetics , Prevotella/pathogenicity , Streptococcus mutans/genetics , Streptococcus mutans/pathogenicity
10.
Sci Rep ; 10(1): 2771, 2020 02 17.
Article in English | MEDLINE | ID: mdl-32066758

ABSTRACT

The presence of advanced fibrosis is an important measure of the severity of chronic liver disease. Prior works that have examined the gut microbiome as a novel biomarker for advanced fibrosis have only examined patients with nonalcoholic fatty liver disease. Therefore, our goal was to examine the gut microbiome across varying etiologies of liver disease to create a predictive model for liver fibrosis based upon a microbial signature. Stool samples were obtained from patients with chronic liver disease (n = 50) undergoing FibroScan (ultrasound elastography) at the VA Greater Los Angeles Healthcare System. Healthy control patients (n = 25) were also recruited as a reference population. Fecal samples underwent 16S ribosomal RNA sequencing. Using differentially abundant microbes, a random forest classifier model was created to distinguish advanced fibrosis from mild/moderate fibrosis. The findings were then validated in a separate cohort of chronic liver disease patients (n = 37). Etiologies for liver disease included non-alcoholic liver disease (58.0%), hepatitis C (26.0%), hepatitis B (10.0%), and alcohol (6.0%). Microbiome composition was distinct in liver patients with advanced fibrosis compared to those with minimal fibrosis and healthy controls (p = 0.003). In multivariate negative binomial modeling, 26 bacterial taxa were differentially abundant in patients with advanced fibrosis as compared to those with minimal/moderate fibrosis (q-value < 0.05). A random forests classifier based on these taxa had an AUROC of 0.90 to predict advanced fibrosis. Prevotella copri, which was enriched in patients with advanced fibrosis, was the most strongly predictive microbe in the classifier. The classifier had an AUROC of 0.82 for advanced fibrosis in the validation cohort and Prevotella copri remained the strongest predictive microbe for advanced fibrosis. There is a distinct microbial signature for patients with advanced fibrosis independent of liver disease etiology and other comorbidities. These results suggest that microbial profiles can be used as a non-invasive marker for advanced fibrosis and support the hypothesis that microbes and their metabolites contribute to hepatic fibrosis.


Subject(s)
End Stage Liver Disease/genetics , Gastrointestinal Microbiome/genetics , Liver Cirrhosis/genetics , Non-alcoholic Fatty Liver Disease/genetics , Adult , Aged , End Stage Liver Disease/complications , End Stage Liver Disease/microbiology , End Stage Liver Disease/pathology , Feces/microbiology , Female , Humans , Liver/microbiology , Liver/pathology , Liver Cirrhosis/complications , Liver Cirrhosis/microbiology , Liver Cirrhosis/pathology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/pathology , Prevotella/isolation & purification , Prevotella/pathogenicity , Severity of Illness Index
11.
J Mol Neurosci ; 70(6): 887-896, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32062762

ABSTRACT

The role of gut microbiome was recently raised in the pathogenesis of neurodevelopmental disorders including autism spectrum disorder (ASD). The aim of this study was to elucidate changes in gut microbiome in Egyptian autistic children and its possible correlation with the severity of autism and gastrointestinal (GI) symptoms. The gut bacterial microbiome of 41 ASD children, 45 siblings, and 45 healthy controls were analyzed using quantitative SYBR Green real-time PCR technique targeting 16S rRNA of selected bacteria. The gut microbiome of ASD children and their siblings contained a higher relative abundance of Bacteroides as well as Ruminococcus than controls. Prevotella/Bacteroides (P/B) ratio and Firmicutes/Bacteroidetes (F/B) were significantly lower in both ASD cases and their siblings. The only difference between the autistic cases and their siblings was the significantly higher level of Bifidobacterium in siblings, which appears to offer them a protective role. There was no correlation between the altered gut microbiome and the severity of autism or GI symptoms. The current study showed an evidence of changes in the gut microbiome of autistic children compared to the unrelated control. However, the microbiome profile of siblings was more like that of autistic children than that of unrelated controls indicating that gut microbiota is affected by dietary habits, living conditions together with host genetic factors.


Subject(s)
Autism Spectrum Disorder/microbiology , Gastrointestinal Microbiome , Bacteroides/genetics , Bacteroides/pathogenicity , Bifidobacterium/genetics , Bifidobacterium/pathogenicity , Child , Child, Preschool , Female , Firmicutes/genetics , Firmicutes/pathogenicity , Humans , Infant , Male , Prevotella/genetics , Prevotella/pathogenicity , RNA, Ribosomal, 16S/genetics , Tertiary Care Centers/statistics & numerical data
12.
Braz J Microbiol ; 51(1): 151-159, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31332740

ABSTRACT

Preterm birth remains the main contributor to early childhood mortality. The vaginal environment, including microbiota composition, might contribute to the risk of preterm delivery. Alterations in the vaginal microbial community structure might represent a risk factor for preterm birth. Here, we aimed to (a) investigate the association between preterm birth and the vaginal microbial community and (b) identify microbial biomarkers for risk of preterm birth. Microbial DNA was isolated from vaginal swabs in a cohort of 69 women enrolled at hospital admission for their delivery. Microbiota was analyzed by high-throughput 16S rRNA sequencing. While no differences in microbial diversity measures appeared associated with the spontaneous preterm and full-term outcomes, the microbial composition was distinct for these groups. Differential abundance analysis showed Lactobacillus species to be associated with full-term birth whereas an unknown Prevotella species was more abundant in the spontaneous preterm group. Although we studied a very miscegenated population from Brazil, our findings were similar to evidence pointed by other studies in different countries. The role of Lactobacillus species as a protector in the vaginal microbiome is demonstrated to be also a protector of spontaneous preterm outcome whereas the presence of pathogenic species, such as Prevotella spp., is endorsed as a factor of risk for spontaneous preterm delivery.


Subject(s)
Bacteria/classification , Microbiota , Obstetric Labor, Premature/diagnosis , Vagina/microbiology , Adult , Bacteria/isolation & purification , Biomarkers/analysis , Brazil , Cohort Studies , Female , High-Throughput Nucleotide Sequencing , Humans , Lactobacillus/classification , Obstetric Labor, Premature/microbiology , Pregnancy , Prevotella/classification , Prevotella/pathogenicity , RNA, Ribosomal, 16S , Risk Factors , Young Adult
13.
J Infect Dis ; 220(9): 1399-1405, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31369673

ABSTRACT

Bacterial vaginosis (BV) is the most common cause of vaginal discharge. It is associated with an increased risk of preterm delivery, pelvic inflammatory disease, and an increased risk of acquisition of sexually transmitted infections including human immunodeficiency virus (HIV). The epidemiology of BV supports sexual transmission. However, its etiology remains unknown. At the center of the debate is whether BV is caused by a primary pathogen or a polymicrobial consortium of microorganisms that are sexually transmitted. We previously published a conceptual model hypothesizing that BV is initiated by sexual transmission of Gardnerella vaginalis. Critics of this model have iterated that G. vaginalis is found in virginal women and in sexually active women with a normal vaginal microbiota. In addition, colonization does not always lead to BV. However, recent advances in BV pathogenesis research have determined the existence of 13 different species within the genus Gardnerella. It may be that healthy women are colonized by nonpathogenic Gardnerella species, whereas virulent strains are involved in BV development. Based on our results from a recent prospective study, in addition to an extensive literature review, we present an updated conceptual model for the pathogenesis of BV that centers on the roles of virulent strains of G. vaginalis, as well as Prevotella bivia and Atopobium vaginae.


Subject(s)
Actinobacteria/growth & development , Gardnerella vaginalis/growth & development , Prevotella/growth & development , Vagina/microbiology , Vaginosis, Bacterial/physiopathology , Actinobacteria/pathogenicity , Female , Gardnerella vaginalis/pathogenicity , Humans , Models, Biological , Prevotella/pathogenicity , Virulence
14.
Biomolecules ; 9(4)2019 04 09.
Article in English | MEDLINE | ID: mdl-30970641

ABSTRACT

Chronic inflammatory autoimmune disorders are systemic diseases with increasing incidence and still lack a cure. More recently, attention has been placed in understanding gastrointestinal (GI) dysbiosis and, although important progress has been made in this area, it is currently unclear to what extent microbiome manipulation can be used in the treatment of autoimmune disorders. Via the use of appropriate models, rheumatoid arthritis (RA), a well-known exemplar of such pathologies, can be exploited to shed light on the currently overlooked effects of existing therapies on the GI microbiome. In this direction, we here explore the crosstalk between the GI microbiome and the host immunity in model arthritis (collagen induced arthritis, CIA). By exploiting omics from samples of limited invasiveness (blood and stools), we assess the host-microbiome responses to standard therapy (methotrexate, MTX) combined with mechanical subcutaneous stimulation (MS) and to mechanical stimulation alone. When MS is involved, results reveal the sphingolipid metabolism as the trait d'union among known hallmarks of (model) RA, namely: Imbalance in the S1P-S1PR1 axis, expansion of Prevotellasp., and invariant Natural Killer T (iNKT)-penia, thus offering the base of a rationale to mechanically modulate this pathway as a therapeutic target in RA.


Subject(s)
Arthritis, Experimental/microbiology , Gastrointestinal Microbiome , Host-Pathogen Interactions , Sphingolipids/metabolism , Animals , Antirheumatic Agents/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Female , Killer Cells, Natural/immunology , Methotrexate/therapeutic use , Prevotella/pathogenicity , Rats , Rats, Wistar , Stress, Mechanical
15.
J Infect Dis ; 219(3): 497-508, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30202890

ABSTRACT

Background: Human immunodeficiency virus (HIV) infection is an independent risk factor for coronary heart disease (CHD) and is associated with perturbation of the gut microbiota. Methods: We analyzed gut microbiota in 30 HIV-infected individuals with CHD (CHD+) and 30 without CHD (CHD-) of the HIV-HEART study group. Results: Gut microbiota linked to CHD was associated with lower α-diversity. Despite insignificant differences in ß-diversity, co-occurrence networks of bacterial genera clearly diverged between CHD+ and CHD- individuals. Multidimensional scaling separated HIV-infected individuals into 2 microbiome clusters, dominated by the genus Prevotella or Bacteroides. The relative abundance of 49 other genera was significantly different between both clusters. The Prevotella-rich cluster was largely composed of men who have sex with men (MSM) (97%), whereas the Bacteroides-rich cluster comprised both MSM (45%) and heterosexual individuals (55%). MSM of the Bacteroides-rich cluster were characterized by reduced α-diversity, advanced immunological HIV stage, longer antiretroviral therapy with more ART regimens, and longer use of protease inhibitors, compared with Prevotella-rich MSM. Conclusions: Community structures of gut microbiota rather than individual species might facilitate risk assessment of CHD in HIV-infected individuals. Sexual behavior appears to be an important factor affecting gut microbiota ß-diversity and should be considered in future studies.


Subject(s)
Biodiversity , Coronary Disease/complications , Gastrointestinal Microbiome , HIV Infections/complications , Adult , Aged , Bacteroides/genetics , Bacteroides/isolation & purification , Bacteroides/pathogenicity , Female , Homosexuality, Male , Humans , Male , Methylamines/pharmacology , Methylamines/therapeutic use , Middle Aged , Prevotella/genetics , Prevotella/isolation & purification , Prevotella/pathogenicity , Risk Factors , Sexual Behavior , Sexual and Gender Minorities
16.
Anaerobe ; 54: 128-135, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30189320

ABSTRACT

Quorum sensing (QS) signaling regulates the motility, adhesion, and biofilm formation of bacteria, and at the same time activates immune response in eukaryotic organisms. We recently demonstrated that the QS molecule, dihydroxy-2, 3-pentanedione (DPD), and its analogs significantly inhibit estradiol-regulated virulence of Prevotella aurantiaca, one of the four species in the Prevotella intermedia group. Here, we examined the combined effects of estradiol and QS signaling on 1) cytokine response of human gingival keratinocytes (HMK) against whole cell extract (WCE) of P. intermedia, Prevotella nigrescens, and Prevotella pallens, and 2) biofilm formation of these three Prevotella species. All experiments were performed in the presence or absence of estradiol, and with different QS molecules: DPD and its analogs (ethyl-DPD, butyl-DPD, and isobutyl-DPD). Concentrations of interleukin (IL)-1ß, -6, and -8 were determined by the Luminex multiplex immunoassay, biofilm mass was quantitatively evaluated by measuring protein concentration via the Bradford method, and the microtopography of biofilms was assessed by scanning electron microscopy (SEM) imaging. Concentrations of IL-6 and IL-8 were elevated when HMK cells were incubated with estradiol and WCE of P. intermedia and P. nigrescens, but decreased when incubated with estradiol and WCE of P. pallens. Butyl-DPD neutralized the estradiol- and WCE-induced regulation of HMK interleukin expression and, at the same time, inhibited the biofilm formation of P. intermedia and P. nigrescens. SEM micrographs revealed a decrease in biofilm mass after application of butyl-DPD, which was most detectable among the P. intermedia ATCC 25611 and P. nigrescens ATCC 33563 and AHN 8293 strains. In conclusion, butyl-DPD analog is able to neutralize the WCE-induced epithelial cytokine response and, at the same time, to inhibit the biofilm formation of P. intermedia and P. nigrescens.


Subject(s)
Bacteroidaceae Infections/immunology , Epithelial Cells/immunology , Gingiva/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Interleukin-8/immunology , Prevotella/physiology , Quorum Sensing , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/microbiology , Biofilms , Epithelial Cells/microbiology , Gingiva/microbiology , Humans , Interleukin-1beta/genetics , Interleukin-6/genetics , Interleukin-8/genetics , Keratinocytes/immunology , Keratinocytes/microbiology , Prevotella/classification , Prevotella/genetics , Prevotella/pathogenicity , Prevotella intermedia/genetics , Prevotella intermedia/pathogenicity , Prevotella intermedia/physiology , Prevotella nigrescens/genetics , Prevotella nigrescens/pathogenicity , Prevotella nigrescens/physiology
17.
Int Endod J ; 51(11): 1196-1204, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29737548

ABSTRACT

AIM: To determine the presence of Prevotella species, the cfxA/cfxA2, blaZ and blaTEM genes associated with resistance to lactamic agents in different oral niches of children with pulp necrosis. METHODOLOGY: Children with pulp necrosis in primary teeth had samples of saliva, supragingival, pulp chamber and root canal biofilms collected and tested for Prevotella species (P. intermedia, P. nigrescens, P. tannerae) and for beta-lactam resistance genes (cfxA/cfxA2, blaZ and blaTEM). The presence of bacterial DNA was examined through PCR, with a specific primer directed to the 16S rRNA gene. Specific primers were used to detect the Prevotella species and beta-lactam resistance genes. The chi-square test was used to analyse associations between the presence of bacteria and clinical variables. The Cochran's Q test was used to assess whether the proportion of gene detection is the same between different sites. RESULTS: Thirty-two teeth were sampled from 27 patients with a mean age of 5.5 years (±1.76). The total detection rate of Prevotella strains was 29.1%, 25%, 21.8% and 32.29% in saliva, supragingival, pulp chamber and root canal samples, respectively. P. nigrescens was the most commonly detected species in all oral niches. The previous use of antibiotics was associated with detection of P. nigrescens in saliva (P = 0.03). Pain was associated with the presence of P. nigrescens (P = 0.04) and P. tannerae (P = 0.01) in pulp chamber biofilm. blaTEM was detected in the four oral niches, being more frequent (23.8%) in supragingival biofilm (Cochran's Q test, P = 0.04). The presence of P. intermedia in SB and PC was associated with the detection of blaTEM in saliva (P = 0.04). The cfxA/cfxA2 and blaZ genes were not detected in any of the four oral niches. CONCLUSIONS: The oral cavity of children with pulp necrosis had a variable distribution of Prevotella strains in different niches. Saliva, supragingival biofilm, pulp chamber and root canals of primary teeth with necrotic pulps can harbour resistance genes to beta-lactams agents.


Subject(s)
Dental Pulp Cavity/microbiology , Dental Pulp Necrosis/microbiology , Lactams/pharmacology , Prevotella/genetics , beta-Lactam Resistance/genetics , Anti-Bacterial Agents/pharmacology , Bacteroidaceae Infections/microbiology , Biofilms , Brazil , Child , Child, Preschool , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Genes, Bacterial/genetics , Humans , Mouth/microbiology , Prevotella/pathogenicity , RNA, Ribosomal, 16S/genetics , Saliva/microbiology , Tooth, Deciduous , beta-Lactamases/genetics
18.
Article in English | MEDLINE | ID: mdl-29439973

ABSTRACT

DS-2969b is a novel GyrB inhibitor in development for the treatment of Clostridium difficile infection (CDI). The aim of this study was to assess the safety, tolerability, pharmacokinetics, and effects on the normal gastrointestinal microbiota of multiple daily oral ascending doses of DS-2969b in healthy subjects. The study enrolled three sequential ascending-dose cohorts (60 mg, 200 mg, and 400 mg). In each cohort, subjects received an oral dose of DS-2969b or placebo (six subjects received DS-2969b, and two received placebo) each morning for 14 days. DS-2969b was safe and well tolerated at all dose levels examined. All adverse events related to DS-2969b were mild and predominantly related to the gastrointestinal tract. DS-2969a (free form of DS-2969b) plasma concentrations increased with increasing doses; however, both the maximum concentration of drug in serum (Cmax) and the area under the concentration-time curve (AUC) increased less than dose proportionally. In all cohorts, sufficient fecal levels of DS-2969a were achieved within 24 h following the administration of the first dose and maintained for at least 17 days. Following treatment with DS-2969b, clear reductions in the populations of Clostridium coccoides and Bifidobacterium groups were observed. However, populations of three other bacterial groups examined (Bacteroides fragilis, Clostridium leptum, and Prevotella) were not affected. Data from this study support and encourage the further development of DS-2969b as a novel treatment for CDI.


Subject(s)
Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacokinetics , Topoisomerase II Inhibitors/pharmacokinetics , Administration, Oral , Adolescent , Adult , Bacteroides fragilis/drug effects , Bacteroides fragilis/metabolism , Bifidobacterium/drug effects , Bifidobacterium/pathogenicity , Clostridioides difficile/drug effects , Clostridioides difficile/pathogenicity , Clostridium Infections/metabolism , Double-Blind Method , Drug Administration Schedule , Female , Gastrointestinal Microbiome/drug effects , Healthy Volunteers , Humans , Male , Middle Aged , Prevotella/drug effects , Prevotella/pathogenicity , Topoisomerase II Inhibitors/adverse effects , Young Adult
20.
Cell Rep ; 20(6): 1269-1277, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28793252

ABSTRACT

The human gut is colonized by a large number of microorganisms (∼1013 bacteria) that support various physiologic functions. A perturbation in the healthy gut microbiome might lead to the development of inflammatory diseases, such as multiple sclerosis (MS). Therefore, gut commensals might provide promising therapeutic options for treating MS and other diseases. We report the identification of human gut-derived commensal bacteria, Prevotella histicola, which can suppress experimental autoimmune encephalomyelitis (EAE) in a human leukocyte antigen (HLA) class II transgenic mouse model. P. histicola suppresses disease through the modulation of systemic immune responses. P. histicola challenge led to a decrease in pro-inflammatory Th1 and Th17 cells and an increase in the frequencies of CD4+FoxP3+ regulatory T cells, tolerogenic dendritic cells, and suppressive macrophages. Our study provides evidence that the administration of gut commensals may regulate a systemic immune response and may, therefore, have a possible role in treatment strategies for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Gastrointestinal Microbiome , Prevotella/immunology , Probiotics/therapeutic use , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/microbiology , Forkhead Transcription Factors/metabolism , Genes, MHC Class II , Humans , Macrophages/immunology , Mice , Prevotella/pathogenicity , Th1 Cells/immunology , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...