Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 919
Filter
1.
Biomed Pharmacother ; 180: 117467, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39362066

ABSTRACT

Commensal microbiota is crucial for nutrient digestion and production of biologically active molecules, many of which mimic endogenous ligands of human GPCRs. Bacteroides spp. are among the most abundant bacteria residing in the human gut and their absence has been positively correlated with metabolic disorders. In the present study, we focused on N-acylated glycines (NAGlys) as products of Bacteroides spp. and potential GPCR ligands modulating GLP-1 secretion. Representative strains of the most abundant commensal Bacteroides were cultured in either yeast- or animal-based nutrient broths. The broths post-culture were investigated in terms of the contents of NAGlys and stimulatory effects towards GLP-1 production in GLUTag and NCI-H716 cell lines. Pure preparations of the detected NAGlys were further studied to evaluate stimulation of GLP-1 production and related cellular signalling evoked. The most potent NAGlys were also tested as ligands of key lipid GPCRs involved in the regulation of carbohydrate metabolism: GPR40/FFAR1, GPR55, GPR119, and GPR120/FFAR4. We found that Bacteroides potentiate GLP-1 production, depending on the strain and provided nutrient mix. Long-chain unsaturated oleoyl and arachidonoyl glycines, produced by B. thetaiotaomicron and B. intestinalis in the animal-based broth, were particularly effective in stimulation of GLP-1 secretion. They served as agonists of all the receptors under study expressed in GLP-1-producing cells. The obtained results broaden the knowledge of microbial signalling molecules and their role in regulation of carbohydrate homeostasis. They also emphasise the importance of balanced diet as a source of building blocks for commensal bacteria to produce efficient agonists of lipid GPCRs.

2.
FEMS Microbes ; 5: xtae027, 2024.
Article in English | MEDLINE | ID: mdl-39391373

ABSTRACT

The gastrointestinal tract (GIT) is typically considered the natural niche of enterococci. However, these bacteria also inhabit extraintestinal tissues, where they can disrupt organ physiology and cause life-threatening infections. Here, we discuss how enterococci, primarily Enterococcus faecalis, interact with the intestine and other host anatomical locations such as the oral cavity, heart, liver, kidney, and vaginal tract. The metabolic flexibility of these bacteria allows them to quickly adapt to new environments, promoting their persistence in diverse tissues. In transitioning from commensals to pathogens, enterococci must overcome harsh conditions such as nutrient competition, exposure to antimicrobials, and immune pressure. Therefore, enterococci have evolved multiple mechanisms to adhere, colonize, persist, and endure these challenges in the host. This review provides a comprehensive overview of how enterococci interact with diverse host cells and tissues across multiple organ systems, highlighting the key molecular pathways that mediate enterococcal adaptation, persistence, and pathogenic behavior.

3.
BMC Oral Health ; 24(1): 1086, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39277730

ABSTRACT

BACKGROUND: This single-blind randomized controlled trial was aimed to evaluate the microbiological and clinical effects of Zeger therapy on gingival health. METHODS: Twenty-four adults with gingivitis were recruited and monitored micro-biologically and clinically at baseline (Day 0), 4 weeks (Day 29) after therapy. All volunteers received one-stage full-mouth supragingival scaling as basic oral health care for baseline, and then randomly divided into experimental (koumiss, n = 12) or control (none, n = 12) group. The koumiss was used once a day for 4 weeks. RESULTS: The microbial diversity of the experimental group increased significantly after drinking koumiss (p < 0.05), mainly owing to increasing of Gram-positive bacteria (p = 0.038) and oral health-related microbes (Rothia, Corynebacterium, Actinomyces, Saccharibacteria_TM7, etc.), decreasing of Gram-negative bacteria (p = 0.009) and periodontal disease-related microbes (Porphyromonas, Fusobacterium, Veillonella, etc.), while the microbial diversity of the control group had no significant change (p > 0.05). However, there was no significant difference between the two groups in the clinical parameters (p > 0.05). CONCLUSIONS: Zeger therapy promotes the diversity of supragingival microbiome in adults with gingivitis and increases the abundance of some beneficial flora while decreasing some harmful without clinical parameters marked changing, which holds promise for improving of gingivitis and may be a valuable oral health care approach in the future. TRIAL REGISTRATION: The clinical trial was approved by the Medical Ethics Committee of West China Hospital of Stomatology, Sichuan University, batch No. WCHSIRB-D-2021-428. Before patient registration began, the prospective clinical trial was registered in www. CLINICALTRIALS: gov public repository in China under the registration number ChiCTR2200060555 on 04/06/2022.


Subject(s)
Gingivitis , Probiotics , Humans , Gingivitis/microbiology , Gingivitis/therapy , Female , Male , Adult , Single-Blind Method , Probiotics/therapeutic use , Young Adult , Middle Aged , Dental Scaling/methods
4.
Indian J Microbiol ; 64(3): 1301-1309, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39282185

ABSTRACT

Staphylococcus aureus possesses sar family genes, including sarA, S, R, T, U, V, X, Y, Z, and rot, which are transcription factors involved in biofilm formation and quorum sensing. In contrast, Staphylococcus epidermidis has sarA, R, V, X, Y, Z, and rot genes; specifically, SarA, Z, and X are involved in biofilm formation. The expression of the sar family members in S. epidermidis isolated from clinical and non-clinical environments is unknown. This study aimed to establish if clinical and non-clinical isolates of S. epidermidis express the sar family members. We genotyped isolates from clinical ocular infections (n = 52), or non-clinical healthy conjunctiva (n = 40), and healthy skin (n = 50), using multilocus sequence typing (MLST) and the staphylococcal chromosomal cassette mec (SCCmec). We selected strains with different genotypes and representatives of each source of isolation, and the presence of the sar family genes was detected using PCR and RT-qPCR to determine their expression. The sar family genes were present in all selected strains, with no observed differences. The relative expression of the sar family showed that all selected strains expressed each gene weakly, with no significant differences observed between them or between different sources of isolation. In conclusion, the presence and relative expression of the sar family genes are very similar among strains, with no differences based on their origin of isolation and genotype. Supplementary Information: The online version contains supplementary material available at 10.1007/s12088-024-01339-x.

5.
Cureus ; 16(8): e67088, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39286694

ABSTRACT

We hope to add to the literature evidence regarding the increasing morbidity associated with an invasive infection by a normal body commensal, Streptococcus constellatus (S. constellatus). An increasing amount of literature documentation of intra- and extracranial disease manifestations following a systemic infection by this micro-organism is noted. We describe the findings of severe, necrotizing right lung disease and possible septic brain emboli in a 54-year-old gentleman in whom microbiological investigations suggest Streptococcal pneumonia, S. constellatus, and mixed anaerobes as possible culprit micro-organisms causing his severe disease state.

6.
Burns Trauma ; 12: tkae037, 2024.
Article in English | MEDLINE | ID: mdl-39224840

ABSTRACT

Background: Calvatia gigantea (CG) is widely used as a traditional Chinese medicine for wound treatment. In this study, we aimed to determine the effects of CG extract (CGE) on diabetic wound healing and the commensal wound microbiome. Method: A wound model was established using leptin receptor-deficient db/db mice, with untreated mice as the control group and CGE-treated mice as the treatment group. The wound healing rate, inflammation and histology were analyzed. Additionally, wound microbiome was evaluated via 16S ribosomal RNA (rRNA) gene sequencing. Results: CGE significantly accelerated the healing of diabetic ulcer wounds, facilitated re-epithelialization, and downregulated the transcription levels of the inflammatory cytokines, interleukin-1ß and tumor necrosis factor-α. Furthermore, CGE treatment positively affected the wound microbiome, promoting diversity of the microbial community and enrichment of Escherichia-Shigella bacteria in the CGE-treated group. Conclusions: Overall, CGE enhanced diabetic wound healing by modulating the wound microbiome and facilitating macrophage polarization during inflammation. These findings suggest modulation of the commensal wound microbiome using medicinal plants as a potential therapeutic strategy for diabetic wounds.

7.
Genes Brain Behav ; 23(5): e70000, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39231190

ABSTRACT

Mutations in voltage-gated sodium (Nav) channels, which are essential for generating and propagating action potentials, can lead to serious neurological disorders, such as epilepsy. However, disease-causing Nav channel mutations do not always result in severe symptoms, suggesting that the disease conditions are significantly affected by other genetic factors and various environmental exposures, collectively known as the "exposome". Notably, recent research emphasizes the pivotal role of commensal bacteria in neural development and function. Although these bacteria typically benefit the nervous system under normal conditions, their impact during pathological states remains largely unknown. Here, we investigated the influence of commensal microbes on seizure-like phenotypes exhibited by paraShu-a gain-of-function mutant of the Drosophila Nav channel gene, paralytic. Remarkably, the elimination of endogenous bacteria considerably ameliorated neurological impairments in paraShu. Consistently, reintroducing bacteria, specifically from the Lactobacillus or Acetobacter genera, heightened the phenotypic severity in the bacteria-deprived mutants. These findings posit that particular native bacteria contribute to the severity of seizure-like phenotypes in paraShu. We further uncovered that treating paraShu with antibiotics boosted Nrf2 signaling in the gut, and that global Nrf2 activation mirrored the effects of removing bacteria from paraShu. This raises the possibility that the removal of commensal bacteria suppresses the seizure-like manifestations through augmented antioxidant responses. Since bacterial removal during development was critical for suppression of adult paraShu phenotypes, our research sets the stage for subsequent studies, aiming to elucidate the interplay between commensal bacteria and the developing nervous system in conditions predisposed to the hyperexcitable nervous system.


Subject(s)
Seizures , Voltage-Gated Sodium Channels , Animals , Seizures/genetics , Voltage-Gated Sodium Channels/genetics , Voltage-Gated Sodium Channels/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/microbiology , Phenotype , Mutation , Acetobacter/genetics , Acetobacter/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Symbiosis/genetics , Gastrointestinal Microbiome
8.
Immunol Rev ; 326(1): 203-218, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39285525

ABSTRACT

The increasing prevalence of immune-mediated non-communicable chronic diseases, such as food allergies, has prompted a deeper investigation into the role of the gut microbiome in modulating immune responses. Here, we explore the complex interactions between commensal microbes and the host immune system, highlighting the critical role of gut bacteria in maintaining immune homeostasis. We examine how modern lifestyle practices and environmental factors have disrupted co-evolved host-microbe interactions and discuss how changes in microbiome composition impact epithelial barrier function, responses to food allergens, and susceptibility to allergic diseases. Finally, we examine the potential of bioengineered microbiome-based therapies, and live biotherapeutic products, for reestablishing immune homeostasis to prevent or treat food allergies.


Subject(s)
Food Hypersensitivity , Gastrointestinal Microbiome , Symbiosis , Humans , Animals , Gastrointestinal Microbiome/immunology , Food Hypersensitivity/immunology , Symbiosis/immunology , Homeostasis , Allergens/immunology , Food , Immunomodulation , Host Microbial Interactions/immunology , Probiotics/therapeutic use
9.
Sci Total Environ ; 954: 176321, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39341232

ABSTRACT

Synanthropic rodents such as the brown rat (Rattus norvegicus) and black rat (Rattus rattus) are a source of disturbance in urban areas and the focus of control programs. Control measures often rely on anticoagulant rodenticides, but their broad use is compromised by the emergence of resistance. Here we studied the prevalence of anticoagulant resistance genotypes in the Vkorc1 gene in rats in the metropolitan area of Barcelona. In this area, part of the management practices to control brown rats include anticoagulant rodenticide use, but rodenticides with different active ingredients are used in rotation. Brown rats were sampled from the sewage system during two periods: from December 2016 to November 2017 when difenacoum and brodifacoum were used, and from August 2021 to July 2022 when bromadiolone was used. Because black rats have just recently been detected in Barcelona, we only studied them during the latter sampling period, with samples obtained from a control action carried out in a green urban area. Exon 3 of the Vkorc1 gene was characterized in both species, while exon 1 was additionally analyzed in black rats. Synonymous mutations, not resulting in amino-acid changes, were found in both exons, indicating no evidence of anticoagulant resistance in the rats of Barcelona. This finding indicates that the current rodent management plan in Barcelona, which involves anticoagulant rotation for brown rats and the use of life capture traps in specific situations for black rats, has helped to prevent the emergence of resistance to anticoagulant rodenticides in rats in Barcelona. Future actions should aim to diversify the control measures included in the rodent management plan.

10.
Microorganisms ; 12(8)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39203490

ABSTRACT

Microbiota present around the ocular surface, encompassing the eyelid skin, the conjunctival sac, and the meibomian glands, play a significant role in various inflammatory conditions associated with the ocular surface. Cutibacterium acnes (C. acnes), formerly, Propionibacterium acnes, is one of the most predominant commensal bacteria and its relative abundance declines with aging. However, it can act as both an infectious and an immunogenic pathogen. As an infectious pathogen, C. acnes has been reported to cause late onset endophthalmitis post-cataract surgery and infectious keratitis. On the other hand, it can trigger immune responses resulting in conditions such as phlyctenules in the cornea, chalazion in the meibomian glands, and granuloma formation in ocular sarcoidosis. This review explores the role of C. acnes in ocular inflammation, specifically highlighting its implications for diagnosis and management.

11.
Dent J (Basel) ; 12(8)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39195076

ABSTRACT

(1) Background: The rise in electronic cigarette (E-cigarette) popularity, especially among adolescents, has prompted research to investigate potential effects on health. Although much research has been carried out on the effect on lung health, the first site exposed to vaping-the oral cavity-has received relatively little attention. The aims of this study were twofold: to examine the effects of E-liquids on the viability and hydrophobicity of oral commensal streptococci, and the effects of E-cigarette-generated aerosols on the biomass and viability of oral commensal streptococci. (2) Methods: Quantitative and confocal biofilm analysis, live-dead staining, and hydrophobicity assays were used to determine the effect on oral commensal streptococci after exposure to E-liquids and/or E-cigarette-generated aerosols. (3) Results: E-liquids and flavors have a bactericidal effect on multispecies oral commensal biofilms and increase the hydrophobicity of oral commensal streptococci. Flavorless and some flavored E-liquid aerosols have a bactericidal effect on oral commensal biofilms while having no effect on overall biomass. (4) Conclusions: These results indicate that E-liquids/E-cigarette-generated aerosols alter the chemical interactions and viability of oral commensal streptococci. Consequently, the use of E-cigarettes has the potential to alter the status of disease and health in the oral cavity and, by extension, affect systemic health.

12.
FEMS Microbiol Ecol ; 100(9)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39169462

ABSTRACT

Candida species primarily exist as harmless commensals in the gastrointestinal tract of warm-blooded animals. However, they can also cause life-threatening infections, which are often associated with gut microbial dysbiosis. Identifying the microbial actors that restrict Candida to commensalism remains a significant challenge. In vitro models could enable a mechanistic study of the interactions between Candida and simulated colon microbiomes. Therefore, this study aimed to elucidate the spatial and temporal colonization kinetics of specific Candida, including C. albicans, C. tropicalis, and C. parapsilosis, and their relative Nakaseomyces glabratus, by using an adapted SHIME® model, simulating the ileum, and proximal and distal colons. We monitored fungal and bacterial colonization kinetics under conditions of eubiosis (commensal lifestyle) and antibiotic-induced dysbiosis (pathogenic lifestyle). Our findings highlighted the variability in the colonization potential of Candida species across different intestinal regions. The ileum compartment proved to be the most favourable environment for C. albicans and C. parapsilosis under conditions of eubiosis. Antibiotic-induced dysbiosis resulted in resurgence of opportunistic Candida species, especially C. tropicalis and C. albicans. Future research should focus on identifying specific bacterial species influencing Candida colonization resistance and explore the long-term effects of antibiotics on the mycobiome and bacteriome.


Subject(s)
Candida , Dysbiosis , Gastrointestinal Microbiome , Ileum , Humans , Candida/drug effects , Candida/growth & development , Ileum/microbiology , Ileum/drug effects , Dysbiosis/microbiology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/adverse effects , Models, Biological , Intestinal Mucosa/microbiology
13.
Front Immunol ; 15: 1415026, 2024.
Article in English | MEDLINE | ID: mdl-39104521

ABSTRACT

Introduction: Galactose-deficient IgA1 (GdIgA1) is critical in the formation of immunodeposits in IgA nephropathy (IgAN), whereas the origin of GdIgA1 is unknown. We focused on the immune response to fecal microbiota in patients with IgAN. Methods: By running 16S ribosomal RNA gene sequencing, we compared IgAN samples to the control samples from household-matched or non-related individuals. Levels of plasma GdIgA1 and poly-IgA complexes were measured, and candidate microbes that can either incite IgA-directed antibody response or degrade IgA through specific IgA protease activities were identified. Results: The IgAN group showed a distinct composition of fecal microbiota as compared to healthy controls. Particularly, high abundance of Escherichia-Shigella was associated with the disease group based on analyses using receiver operating characteristic (area under curve, 0.837; 95% CI, 0.738-0.914), principle coordinates, and the linear discriminant analysis effect size algorithm (linear discriminant analysis score, 4.56; p < 0.001). Accordingly, the bacterial levels directly correlated with high titers of plasma GdIgA1(r = 0.36, p < 0.001), and patients had higher IgA1 against stx2(2.88 ± 0.46 IU/mL vs. 1.34 ± 0.35 IU/mL, p = 0.03), the main antigen of Escherichia-Shigella. Conversely, the healthy controls showed relatively higher abundance of the commensal bacteria that produce IgA-degrading proteases. Particularly, the abundance of some intestinal bacteria expressing IgA proteases showed an inverse correlation with the levels of plasma GdIgA1 in IgAN. Conclusion: Our data suggest that mucosal IgA production, including those of GdIgA1, is potentially linked to the humoral response to gut Escherichia-Shigella as one of the sources of plasma GdIgA1. Conversely, the IgA protease-producing microbiota in the gut are suppressed in patients with IgAN.


Subject(s)
Galactose , Gastrointestinal Microbiome , Glomerulonephritis, IGA , Immunity, Humoral , Immunoglobulin A , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/microbiology , Humans , Gastrointestinal Microbiome/immunology , Immunoglobulin A/immunology , Immunoglobulin A/blood , Male , Female , Adult , Feces/microbiology , Middle Aged , RNA, Ribosomal, 16S/genetics
14.
J Hazard Mater ; 478: 135526, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39153300

ABSTRACT

Phenol and p-cresol are two common toxic small molecules related to various diseases. Existing reports confirmed that high L-tyrosine in the daily diet can increase the concentration of phenolic compounds in blood and urine. L-tyrosine is a common component of protein-rich foods. Some anaerobic bacteria in the gut can convert non-toxic l-tyrosine into these two toxic phenolic compounds, phenol and p-cresol. Existing methods have been constructed for measuring the concentration of phenolic compound in feces. However, there is still a lack of direct visual evidence to measure the phenolic compounds in the intestine. In this study, we aimed to construct a whole-cell biosensor for phenolic compounds detection based on the dmpR, the regulator from the phenol metabolism cluster. The commensal bacterium Citrobacter amalonaticus PS01 was selected and used as the chassis. Compared with the biosensor based on ECN1917, the biosensor PS01[dmpR] could better implant into the mouse gut through gavage and showed a higher sensitive to phenolic compound. And the concentration of phenolic compounds in the intestines could be observed with the help of in vivo imaging system using PS01[dmpR]. This paper demonstrated endogenous phenol synthesis in the gut and the strategy of using commensal bacteria to construct whole-cell biosensors for detecting small molecule compounds in the intestines.


Subject(s)
Biosensing Techniques , Intestines , Animals , Citrobacter/metabolism , Cresols/metabolism , Cresols/toxicity , Phenols/toxicity , Mice , Phenol/analysis , Phenol/toxicity , Tyrosine/metabolism
15.
mSphere ; 9(9): e0020424, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39140737

ABSTRACT

We investigated the influence of a Wnt5A-gut microbiota axis on gut B-cell repertoire and protection from infection, having previously demonstrated that Wnt5A in association with gut commensals helps shape gut T-cell repertoire. Accordingly, Wnt5A heterozygous mice, which express less than wild-type level of Wnt5A, and their isolated Peyer's patches (PPs) were studied in comparison with the wild-type counterparts. The percentages of IgM- and IgA-expressing B cells were quite similar in the PP of both sets of mice. However, the PP of the Wnt5A heterozygous mice harbored significantly higher than wild-type levels of microbiota-bound B cell-secreted IgA, indicating the prevalence of a microbial population therein, which is significantly altered from that of wild-type. Additionally, the percentage of PP IgG1-expressing B cells was appreciably depressed in the Wnt5A heterozygous mice in comparison to wild-type. Wnt5A heterozygous mice, furthermore, exhibited notably higher than the wild-type levels of morbidity and mortality following infection with Salmonella typhimurium, a common gut pathogen. Differences in morbidity/mortality correlated with considerable disparity between the PP-B-cell repertoires of the Salmonella-infected Wnt5A heterozygous and wild-type mice, in which the percentage of IgG1-expressing B1b cells in the PP of heterozygous mice remains significantly low as compared to wild-type. Overall, these results suggest that a gut Wnt5A-microbiota axis is intrinsically associated with the maintenance of gut B-cell repertoire and protection from infection.IMPORTANCEAlthough it is well accepted that B cells and microbiota are required for protection from infection and preservation of gut health, a lot remains unknown about how the optimum B-cell repertoire and microbiota are maintained in the gut. The importance of this study lies in the fact that it unveils a potential role of a growth factor termed Wnt5A in the safeguarding of the gut B-cell population and microbiota, thereby protecting the gut from the deleterious effect of infections by common pathogens. Documentation of the involvement of a Wnt5A-microbiota axis in the shaping of a protective gut B-cell repertoire, furthermore, opens up new avenues of investigations for understanding gut disorders related to microbial dysbiosis and B-cell homeostasis that, till date, are considered incurable.


Subject(s)
B-Lymphocytes , Gastrointestinal Microbiome , Wnt-5a Protein , Animals , Wnt-5a Protein/genetics , Wnt-5a Protein/immunology , Gastrointestinal Microbiome/immunology , Mice , B-Lymphocytes/immunology , Peyer's Patches/immunology , Peyer's Patches/microbiology , Salmonella typhimurium/immunology , Salmonella typhimurium/genetics , Salmonella Infections/immunology , Salmonella Infections/microbiology , Mice, Inbred C57BL , Female , Male , Immunoglobulin A/immunology , Immunoglobulin G/immunology
16.
Immunity ; 57(8): 1893-1907.e6, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39096910

ABSTRACT

Naive CD4+ T cells in specific pathogen-free (SPF) mice are characterized by transcriptional heterogeneity and subpopulations distinguished by the expression of quiescence, the extracellular matrix (ECM) and cytoskeleton, type I interferon (IFN-I) response, memory-like, and T cell receptor (TCR) activation genes. We demonstrate that this constitutive heterogeneity, including the presence of the IFN-I response cluster, is commensal independent insofar as being identical in germ-free and SPF mice. By contrast, Nippostrongylus brasiliensis infection altered this constitutive heterogeneity. Naive T cell-intrinsic transcriptional changes acquired during helminth infection correlated with and accounted for decreased immunization response to an unrelated antigen. These compositional and functional changes were dependent variables of helminth infection, as they disappeared at the established time point of its clearance in mice. Collectively, our results indicate that the naive T cell pool is subject to dynamic transcriptional changes in response to certain environmental cues, which in turn permutes the magnitude of the immune response.


Subject(s)
CD4-Positive T-Lymphocytes , Nippostrongylus , Animals , Mice , CD4-Positive T-Lymphocytes/immunology , Nippostrongylus/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Specific Pathogen-Free Organisms , Transcription, Genetic , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Helminthiasis/immunology , Interferon Type I/metabolism , Interferon Type I/immunology , Mice, Inbred C57BL , Lymphocyte Activation/immunology
17.
mSystems ; 9(9): e0067424, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39166876

ABSTRACT

Haemophilus parainfluenzae (Hp) is a Gram-negative, highly prevalent, and abundant commensal in the human oral cavity, and an infrequent extraoral opportunistic pathogen. Hp occupies multiple niches in the oral cavity, including the supragingival plaque biofilm. Little is known about how Hp interacts with its neighbors in healthy biofilms nor its mechanisms of pathogenesis as an opportunistic pathogen. To address this, we identified the essential genome and conditionally essential genes in in vitro biofilms aerobically and anaerobically. Using transposon insertion sequencing (TnSeq) with a highly saturated mariner transposon library in two strains, the ATCC33392 type-strain (Hp 392) and oral isolate EL1 (Hp EL1), we show that the essential genomes of Hp 392 and Hp EL1 are composed of 395 (20%) and 384 (19%) genes, respectively. The core essential genome, consisting of 341 (17%) essential genes conserved between both strains, was composed of genes associated with genetic information processing, carbohydrate, protein, and energy metabolism. We also identified conditionally essential genes for aerobic and anaerobic biofilm growth, which were associated with carbohydrate and energy metabolism in both strains. RNAseq analysis determined that most genes upregulated during anaerobic growth are not essential for Hp 392 anaerobic survival. The completion of this library and analysis under these conditions gives us a foundational insight into the basic biology of H. parainfluenzae in differing oxygen conditions, similar to its in vivo habitat. This library presents a valuable tool for investigation into conditionally essential genes for an organism that lives in close contact with many microbial species in the human oral habitat.IMPORTANCEHaemophilus parainfluenzae is a highly abundant human commensal microbe, present in most healthy individuals where it colonizes the mouth. H. parainfluenzae correlates with good oral health and may play a role in preservation of healthy host status. Also, H. parainfluenzae can cause opportunistic infections outside of the oral cavity. To date, little is known about how H. parainfluenzae colonizes the human host, despite being such a frequent and abundant part of our human microbiome. Here, we demonstrate the creation and use of a powerful tool, a TnSeq library, used to identify genes necessary for both the outright growth of this organism and also genes conditionally essential for growth in varying oxygen status which it can encounter in the human host. This tool and these data serve as a foundation for further study of this relatively unknown organism that may play a role in preserving human health.


Subject(s)
Biofilms , Genes, Essential , Haemophilus parainfluenzae , Biofilms/growth & development , Haemophilus parainfluenzae/genetics , Genes, Essential/genetics , Humans , Genome, Bacterial/genetics , DNA Transposable Elements/genetics , Microbial Viability/genetics
18.
Crit Rev Microbiol ; : 1-12, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39132685

ABSTRACT

Dental caries, as a biofilm-related disease, is closely linked to dysbiosis in microbial ecology within dental biofilms. Beyond its impact on oral health, bacteria within the oral cavity pose systemic health risks by potentially entering the bloodstream, thereby increasing susceptibility to bacterial endocarditis, among other related diseases. Streptococcus mutans, a principal cariogenic bacterium, possesses virulence factors crucial to the pathogenesis of dental caries. Its ability to adhere to tooth surfaces, produce glucans for biofilm formation, and metabolize sugars into lactic acid contributes to enamel demineralization and the initiation of carious lesions. Its aciduricity and ability to produce bacteriocins enable a competitive advantage, allowing it to thrive in acidic environments and dominate in changing oral microenvironments. In contrast, commensal streptococci, such as Streptococcus sanguinis, Streptococcus gordonii, and Streptococcus salivarius, act as primary colonizers and compete with S. mutans for adherence sites and nutrients during biofilm formation. This competition involves the production of alkali, peroxides, and antibacterial substances, thereby inhibiting S. mutans growth and maintaining microbial balance. This dynamic interaction influences the balance of oral microbiota, with disruptions leading to shifts in microbial composition that are marked by rapid increases in S. mutans abundance, contributing to the onset of dental caries. Thus, understanding the dynamic interactions between commensal and pathogenic bacteria in oral microecology is important for developing effective strategies to promote oral health and prevent dental caries. This review highlights the roles and competitive interactions of commensal bacteria and S. mutans in oral microecology, emphasizing the importance of maintaining oral microbial balance for health, and discusses the pathological implications of perturbations in this balance.

19.
Parasit Vectors ; 17(1): 330, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103931

ABSTRACT

BACKGROUND: Aedes albopictus is an important vector for pathogens such as dengue, Zika, and chikungunya viruses. While insecticides is the mainstay for mosquito control, their widespread and excessive use has led to the increased resistance in Ae. albopictus globally. Gut symbiotic bacteria are believed to play a potential role in insect physiology, potentially linking to mosquitoes' metabolic resistance against insecticides. METHODS: We investigated the role of symbiotic bacteria in the development of resistance in Ae. albopictus by comparing gut symbiotic bacteria between deltamethrin-sensitive and deltamethrin-resistant populations. Adults were reared from field-collected larvae. Sensitive and resistant mosquitoes were screened using 0.03% and 0.09% deltamethrin, respectively, on the basis of the World Health Organization (WHO) tube bioassay. Sensitive and resistant field-collected larvae were screened using 5 × LC50 (lethal concentration at 50% mortality) and 20 × LC50 concentration of deltamethrin, respectively. Laboratory strain deltamethrin-sensitive adults and larvae were used as controls. The DNA of gut samples from these mosquitoes were extracted using the magnetic bead method. Bacterial 16S rDNA was sequenced using BGISEQ method. We isolated and cultured gut microorganisms from adult and larvae mosquitoes using four different media: Luria Bertani (LB), brain heart infusion (BHI), nutrient agar (NA), and salmonella shigella (SS). RESULTS: Sequencing revealed significantly higher gut microbial diversity in field-resistant larvae compared with field-sensitive and laboratory-sensitive larvae (P < 0.01). Conversely, gut microorganism diversity in field-resistant and field-sensitive adults was significantly lower compared with laboratory-sensitive adults (P < 0.01). At the species level, 25 and 12 bacterial species were isolated from the gut of field resistant larvae and adults, respectively. The abundance of Flavobacterium spp., Gemmobacter spp., and Dysgonomonas spp. was significantly higher in the gut of field-resistant larvae compared with sensitive larvae (all P < 0.05). Furthermore, the abundance of Flavobacterium spp., Pantoea spp., and Aeromonas spp. was significantly higher in the gut of field-resistant adults compared with sensitive adults (all P < 0.05). The dominant and differentially occurring microorganisms were also different between resistant larval and adult mosquitoes. These findings suggest that the gut commensal bacteria of Ae. albopictus adults and larvae may play distinct roles in their deltamethrin resistance. CONCLUSIONS: This study provides an empirical basis for further exploration of the mechanisms underlying the role of gut microbial in insecticide resistance, potentially opening a new prospect for mosquito control strategies.


Subject(s)
Aedes , Bacteria , Insecticide Resistance , Insecticides , Larva , Nitriles , Pyrethrins , RNA, Ribosomal, 16S , Symbiosis , Animals , Pyrethrins/pharmacology , Nitriles/pharmacology , Aedes/microbiology , Aedes/drug effects , Insecticides/pharmacology , Larva/microbiology , Larva/drug effects , RNA, Ribosomal, 16S/genetics , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/classification , Gastrointestinal Microbiome/drug effects , Mosquito Vectors/microbiology , Mosquito Vectors/drug effects , DNA, Ribosomal/genetics , Female , DNA, Bacterial/genetics , Gastrointestinal Tract/microbiology
20.
Article in English | MEDLINE | ID: mdl-39110329

ABSTRACT

L-asparaginase is an FDA-approved drug for treating blood cancer, but its inherent antigenicity and L-glutaminase activity are associated with hypersensitivity and organ toxicity. Extracellularly produced glutaminase-free L-asparaginase from human commensal bacteria may be a good alternative to reduce the side effects of therapeutic L-asparaginase. Here, we report the isolation and characterization of fourteen L-asparaginase-producing bacterial strains belonging to the genera Acinetobacter, Escherichia, Klebsiella, and Pseudomonas from human stool and saliva samples. To the best of our knowledge, this is the first report of L-asparaginase-producing human commensal bacterial strains isolated from healthy individuals. L-asparaginase produced by fecal and salivary isolates exhibited significantly higher activity (3.64 to 16.96 U/ml) toward L-asparagine than L-glutamine. Interestingly, L-asparaginase from fecal isolates, Escherichia coli strains 3F1 and 3F2 and salivary isolate Klebsiella pneumoniae 3S3, exhibited no L-glutaminase activity. These isolates were also sensitive to all tested antibiotics. Additionally, these three isolates demonstrated tolerance to pH 3.0 (≥ 88% survival) and 0.3% bile (≥ 95% survival), indicating their potential as probiotics. Among these isolates, L-asparaginase from the highest-producing K. pneumoniae 3S3 strain was found to be a homodimer, with native and subunit molecular weights of 110 kDa and 55 kDa, respectively. The purified enzyme can be further explored for its antitumor and immunomodulatory properties. Overall, future research can be expanded to include the use of a pool of human commensal bacteria as genuine and alternative sources of L-asparaginase for effective cancer treatments and cutting-edge next-generation probiotics.

SELECTION OF CITATIONS
SEARCH DETAIL