Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.158
Filter
1.
Vet Immunol Immunopathol ; 271: 110742, 2024 May.
Article in English | MEDLINE | ID: mdl-38547603

ABSTRACT

Probiotics as dietary additives can improve weight gain, feed efficiency, and disease resistance in cultured fish. In this research, we evaluated and compared the effects of Bacillus subtilis on immunity, mucosal tissue morphology, immune-related gene transcriptions, and intestinal microbiota in flounder (Paralichthys olivaceus) by a 30-day feeding experiment based on a continuous feeding schedule (E1) and a discontinuous feeding schedule (E2). As a result, the use of B. subtilis exerted the best positive effects on survival rate, enzyme activity, mucosal tissue morphology, immune-related gene transcriptions, and intestinal microbiota in flounders. Alkaline phosphatase (AKP), lysozyme (LZM), and superoxide dismutase (SOD) activities in the liver of E2 were higher than those of E1 (P < 0.05). Furthermore, the villi length in the intestinal tract and the fold length in the stomach of E2 were also higher than in E1 (P < 0.05). The il-1 expression levels in the spleen were significantly increased in E2 (P < 0.05) compared to E1. We performed 16 S rRNA sequencing analysis to find that Bacillus in E1 (1.06%) and E2 (1.01%) had higher relative abundances than in E0 (0.053%) at the end of the experiments, indicating that short-term application of B. subtilis with the continuous or discontinuous feeding method can allow both the adaptation of the ecosystem to the presence of probiotics by the establishment of new species in the gut microbiota and the ability these new probiotic species to perform corresponding functions. No significant differences in the ability of probiotic establishment were observed between E1 and E2. Our findings provided a unique perspective to explore the mechanism of immune enhancement with probiotics and to screen the optimal administration strategy in aquaculture application for probiotic use. Together, these results point to some level of enhancement in immune status by continuous and discontinuous feeding after a short-term feeding period, which could be used as a prophylactic strategy for flounder health management.


Subject(s)
Animal Feed , Bacillus subtilis , Flounder , Gastrointestinal Microbiome , Probiotics , Animals , Probiotics/administration & dosage , Probiotics/pharmacology , Flounder/immunology , Flounder/microbiology , Animal Feed/analysis , Feeding Methods/veterinary , Mucous Membrane/immunology , Mucous Membrane/microbiology , Transcription, Genetic
2.
Commun Biol ; 7(1): 171, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38347162

ABSTRACT

Microbial communities at the airway mucosal barrier are conserved and highly ordered, in likelihood reflecting co-evolution with human host factors. Freed of selection to digest nutrients, the airway microbiome underpins cognate management of mucosal immunity and pathogen resistance. We show here the initial results of systematic culture and whole-genome sequencing of the thoracic airway bacteria, identifying 52 novel species amongst 126 organisms that constitute 75% of commensals typically present in heathy individuals. Clinically relevant genes encode antimicrobial synthesis, adhesion and biofilm formation, immune modulation, iron utilisation, nitrous oxide (NO) metabolism and sphingolipid signalling. Using whole-genome content we identify dysbiotic features that may influence asthma and chronic obstructive pulmonary disease. We match isolate gene content to transcripts and metabolites expressed late in airway epithelial differentiation, identifying pathways to sustain host interactions with microbiota. Our results provide a systematic basis for decrypting interactions between commensals, pathogens, and mucosa in lung diseases of global significance.


Subject(s)
Bacteria , Mucous Membrane , Humans , Mucous Membrane/microbiology , Bacteria/genetics , Symbiosis , Immunity, Mucosal , Genomics
3.
Front Cell Infect Microbiol ; 12: 856962, 2022.
Article in English | MEDLINE | ID: mdl-35774401

ABSTRACT

The mucosal surfaces that form the boundary between the external environment and the underlying tissue are protected by a mucus barrier. Mucin glycoproteins, both secreted and cell surface mucins, are the major components of the barrier. They can exclude pathogens and toxins while hosting the commensal bacteria. In this review, we highlight the dynamic function of the mucins and mucus during infection, how this mucosal barrier is regulated, and how pathogens have evolved mechanisms to evade this defence system.


Subject(s)
Mucins , Mucus , Bacteria/metabolism , Glycoproteins/metabolism , Mucins/metabolism , Mucous Membrane/microbiology , Mucus/metabolism
4.
PLoS Pathog ; 18(5): e1010497, 2022 05.
Article in English | MEDLINE | ID: mdl-35580146

ABSTRACT

The mechanisms used by human adapted commensal Neisseria to shape and maintain a niche in their host are poorly defined. These organisms are common members of the mucosal microbiota and share many putative host interaction factors with Neisseria meningitidis and Neisseria gonorrhoeae. Evaluating the role of these shared factors during host carriage may provide insight into bacterial mechanisms driving both commensalism and asymptomatic infection across the genus. We identified host interaction factors required for niche development and maintenance through in vivo screening of a transposon mutant library of Neisseria musculi, a commensal of wild-caught mice which persistently and asymptomatically colonizes the oral cavity and gut of CAST/EiJ and A/J mice. Approximately 500 candidate genes involved in long-term host interaction were identified. These included homologs of putative N. meningitidis and N. gonorrhoeae virulence factors which have been shown to modulate host interactions in vitro. Importantly, many candidate genes have no assigned function, illustrating how much remains to be learned about Neisseria persistence. Many genes of unknown function are conserved in human adapted Neisseria species; they are likely to provide a gateway for understanding the mechanisms allowing pathogenic and commensal Neisseria to establish and maintain a niche in their natural hosts. Validation of a subset of candidate genes confirmed a role for a polysaccharide capsule in N. musculi persistence but not colonization. Our findings highlight the potential utility of the Neisseria musculi-mouse model as a tool for studying the pathogenic Neisseria; our work represents a first step towards the identification of novel host interaction factors conserved across the genus.


Subject(s)
DNA Transposable Elements , Host Microbial Interactions , Neisseria , Animals , Carrier State/microbiology , Carrier State/physiopathology , DNA Transposable Elements/genetics , Gene Library , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Mice , Microbiota/genetics , Mucous Membrane/microbiology , Neisseria/genetics , Neisseria/pathogenicity , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/pathogenicity , Neisseria meningitidis/genetics , Neisseria meningitidis/pathogenicity , Symbiosis/genetics , Symbiosis/physiology , Virulence Factors/genetics
5.
Science ; 376(6596): 950-955, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35617395

ABSTRACT

Associations between the dynamic community of microbes (the microbiota) and the host they colonize appear to be vital for ensuring host health. Microbe-host communication is actively maintained across physiological barriers of various body sites and is mediated by a range of bidirectional secreted proteins and small molecules. So far, a range of "omics" methods have succeeded in revealing the multiplicity of associations between members of a microbiota and a wide range of host processes and diseases. Although these advances point to possibilities for treating disease, there has not been much translational success thus far. We know little about which organisms are key contributors to host health, the importance of strain differences, and the activities of much of the chemical "soup" that is produced by the microbiota. Adding to this complexity are emerging hints of the role of interkingdom interactions between bacteria, phages, protozoa, and/or fungi in regulating the microbiota-host interactions. Functional approaches, although experimentally challenging, could be the next step to unlocking the power of the microbiota.


Subject(s)
Gastrointestinal Microbiome , Host Microbial Interactions , Animals , Humans , Immunity, Mucosal , Mucous Membrane/immunology , Mucous Membrane/microbiology
6.
Mar Biotechnol (NY) ; 24(2): 366-379, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35303209

ABSTRACT

Microbiome plays key roles in the digestion, metabolism, and immunity of the grass carp (Ctenopharyngodon idella). Here, we characterized the normal microbiome of the intestinal contents (IC), skin mucus (SM), oral mucosa (OM), and gill mucosa (GM) of the grass carp, as well as the microbiome of the sidewall (SW) of the raising pool, using full-length 16S rRNA sequencing based on the PacBio platform in this specie for the first time. Twenty phyla, 38 classes, 130 families, 219 genera, and 291 species were classified. One hundred four common classified species might be core microbiota of grass carp. Proteobacteria, Bacteroides, and Cyanobacteria were the dominant phyla in the niche of grass carp. Proteobacteria and Bacteroides dominated the taxonomic composition in the SM, GM, and OM, while Proteobacteria, Planctomycetota, and Cyanobacteria preponderated in the IC and SW groups. Microbiota of IC exhibited higher alpha diversity indices. The microbial communities clustered either in SW or the niche from grass carp, significantly tighter in the SW, based on Bray-Curtis distances (P < 0.05). SM, GM, and OM were similar in microbial composition but were significantly different from IC and SW, while IC had similarity with SW due to their common Cyanobacteria (P < 0.05). Differences were also reflected by niche-specific and differentially abundant microorganisms such as Noviherbaspirillum in the SM and Rhodopseudomonas palustris, Mycobacterium fortuitum, and Acinetobacter schindleri in GM. Significantly raised gene expression was found in IC and SM associated with cell cycle control, cell division, chromosome, coenzyme transport and metabolism, replication, recombination and repair, cell motility, post-translational modification, signal transduction mechanisms, intracellular trafficking, secretion, and vesicles by PICRUSt. This work may be of great value for understanding of fish-microbial co-workshops, especially in different niche of grass carp.


Subject(s)
Carps , Microbiota , Mucous Membrane , Animals , Carps/microbiology , Mucous Membrane/microbiology , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
7.
Sci Immunol ; 7(67): eabe8931, 2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35030034

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are innate-like T lymphocytes that aid in protection against bacterial pathogens at mucosal surfaces through the release of inflammatory cytokines and cytotoxic molecules. Recent evidence suggests that MAIT cells can also provide B cell help. In this study, we describe a population of CXCR5+ T follicular helper (Tfh)­like MAIT cells (MAITfh) that have the capacity to provide B cell help within mucosal lymphoid organs. MAITfh cells are preferentially located near germinal centers in human tonsils and express the classical Tfh-associated transcription factor, B cell lymphoma 6 (BCL-6), the costimulatory markers inducible T cell costimulatory (ICOS) and programmed death receptor 1 (PD-1), and interleukin-21 (IL-21). We demonstrate the ability of MAIT cells to provide B cell help in vivo after mucosal challenge with Vibrio cholerae. Specifically, we show that adoptive transfer of MAIT cells into αß T cell­deficient mice promoted B cell differentiation and increased serum V. cholerae­specific IgA responses. Our data demonstrate the capacity of MAIT cells to participate in adaptive immune responses and suggest that MAIT cells may be potential targets for mucosal vaccines.


Subject(s)
Antibodies/immunology , B-Lymphocytes/immunology , Mucosal-Associated Invariant T Cells/immunology , Mucous Membrane/immunology , Adolescent , Adult , Animals , Antibody Formation/immunology , Child , Child, Preschool , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucous Membrane/microbiology , Vibrio cholerae/immunology
8.
Cell Rep ; 38(1): 110187, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986345

ABSTRACT

Candida albicans is both a commensal and an opportunistic fungal pathogen. Invading hyphae of C. albicans secrete candidalysin, a pore-forming peptide toxin. To prevent cell death, epithelial cells must protect themselves from direct damage induced by candidalysin and by the mechanical forces exerted by expanding hyphae. We identify two key Ca2+-dependent repair mechanisms employed by epithelial cells to withstand candidalysin-producing hyphae. Using camelid nanobodies, we demonstrate candidalysin secretion directly into the invasion pockets induced by elongating C. albicans hyphae. The toxin induces oscillatory increases in cytosolic [Ca2+], which cause hydrolysis of PtdIns(4,5)P2 and loss of cortical actin. Epithelial cells dispose of damaged membrane regions containing candidalysin by an Alg-2/Alix/ESCRT-III-dependent blebbing process. At later stages, plasmalemmal tears induced mechanically by invading hyphae are repaired by exocytic insertion of lysosomal membranes. These two repair mechanisms maintain epithelial integrity and prevent mucosal damage during both commensal growth and infection by C. albicans.


Subject(s)
Candida albicans/metabolism , Candidiasis/pathology , Endosomal Sorting Complexes Required for Transport/metabolism , Fungal Proteins/metabolism , Lysosomes/metabolism , Mucous Membrane/physiology , Animals , Calcium/metabolism , Cell Line , Cell Membrane/physiology , Epithelial Cells/metabolism , Exocytosis/physiology , Fungal Proteins/genetics , Host-Pathogen Interactions , Humans , Hyphae/growth & development , Mice , Mucous Membrane/cytology , Mucous Membrane/microbiology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RAW 264.7 Cells
9.
Microbiol Spectr ; 9(3): e0105821, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34908468

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Hemolysin Proteins/metabolism , Mucous Membrane/microbiology , Streptococcus agalactiae/metabolism , Vagina/microbiology , Animals , Bacterial Adhesion/physiology , Bacterial Proteins/genetics , Bacterial Toxins/metabolism , Cell Line , Epithelial Cells/microbiology , Female , Gene Deletion , Hemolysin Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Pregnancy , Sphingomyelin Phosphodiesterase/metabolism , Streptococcus agalactiae/genetics , Streptococcus agalactiae/growth & development
10.
Front Immunol ; 12: 750808, 2021.
Article in English | MEDLINE | ID: mdl-34917075

ABSTRACT

Endometrial immune response is highly associated with the homeostatic balance of the uterus and embryo development; however, the underlying molecular regulatory mechanisms are not fully elucidated. Herein, the porcine endometrium showed significant variation in mucosal immunity in proliferative and secretory phases by single-cell RNA sequencing. The loose arrangement and high motility of the uterine epithelium in the proliferative phase gave opportunities for epithelial cells and dendritic cells to cross talk with colonizing microbial community, guiding lymphocyte migration into the mucosal and glandular epithelium. The migrating lymphocytes were primarily NK and CD8+ T cells, which were robustly modulated by the chemokine signaling. In the secretory phase, the significantly strengthened mechanical mucosal barrier and increased immunoglobulin A alleviated the migration of lymphocytes into the epithelium when the neuro-modulation, mineral uptake, and amino acid metabolism were strongly upregulated. The noticeably increased intraepithelial lymphocytes were positively modulated by the bacteria in the uterine cavity. Our findings illustrated that significant mucosal immunity variation in the endometrium in the proliferative and secretory phases was closely related to intraepithelial lymphocyte migration, which could be modulated by the colonizing bacteria after cross talk with epithelial cells with higher expressions of chemokine.


Subject(s)
Endometrium/immunology , Endometrium/microbiology , Estrous Cycle/immunology , Immunity, Mucosal/physiology , Microbiota/immunology , Animals , Female , Mucous Membrane/immunology , Mucous Membrane/microbiology , Swine
11.
PLoS Pathog ; 17(12): e1009592, 2021 12.
Article in English | MEDLINE | ID: mdl-34852011

ABSTRACT

Neisseria gonorrhoeae (GC) establishes infection in women from the cervix, lined with heterogeneous epithelial cells from non-polarized stratified at the ectocervix to polarized columnar at the endocervix. We have previously shown that GC differentially colonize and transmigrate across the ecto and endocervical epithelia. However, whether and how GC invade into heterogeneous cervical epithelial cells is unknown. This study examined GC entry of epithelial cells with various properties, using human cervical tissue explant and non-polarized/polarized epithelial cell line models. While adhering to non-polarized and polarized epithelial cells at similar levels, GC invaded into non-polarized more efficiently than polarized epithelial cells. The enhanced GC invasion in non-polarized epithelial cells was associated with increased ezrin phosphorylation, F-actin and ezrin recruitment to GC adherent sites, and the elongation of GC-associated microvilli. Inhibition of ezrin phosphorylation inhibited F-actin and ezrin recruitment and microvilli elongation, leading to a reduction in GC invasion. The reduced GC invasion in polarized epithelial cells was associated with non-muscle myosin II-mediated F-actin disassembly and microvilli denudation at GC adherence sites. Surprisingly, intraepithelial GC were only detected inside epithelial cells shedding from the cervix by immunofluorescence microscopy, but not significantly in the ectocervical and the endocervical regions. We observed similar ezrin and F-actin recruitment in exfoliated cervical epithelial cells but not in those that remained in the ectocervical epithelium, as the luminal layer of ectocervical epithelial cells expressed ten-fold lower levels of ezrin than those beneath. However, GC inoculation induced F-actin reduction and myosin recruitment in the endocervix, similar to what was seen in polarized epithelial cells. Collectively, our results suggest that while GC invade non-polarized epithelial cells through ezrin-driven microvilli elongation, the apical polarization of ezrin and F-actin inhibits GC entry into polarized epithelial cells.


Subject(s)
Cell Polarity , Cytoskeletal Proteins/metabolism , Gonorrhea/microbiology , Neisseria gonorrhoeae/genetics , Actins/metabolism , Cervix Uteri/microbiology , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Epithelium/microbiology , Female , Humans , Microvilli/ultrastructure , Mucous Membrane/microbiology , Neisseria gonorrhoeae/physiology , Phosphorylation
12.
Front Immunol ; 12: 730986, 2021.
Article in English | MEDLINE | ID: mdl-34594336

ABSTRACT

The standard treatment for bacterial vaginosis (BV) with oral metronidazole is often ineffective, and recurrence rates are high among African women. BV-associated anaerobes are closely associated with genital inflammation and HIV risk, which underscores the importance of understanding the interplay between vaginal microbiota and genital inflammation in response to treatment. In this cohort study, we therefore investigated the effects of metronidazole treatment on the vaginal microbiota and genital cytokines among symptomatic South African women with BV [defined as Nugent score (NS) ≥4] using 16S rRNA gene sequencing and multiplex bead arrays. Among 56 BV-positive women, we observed short-term BV clearance (NS <4) in a proportion of women six weeks after metronidazole treatment, with more than half of these experiencing recurrence by 12 weeks post-treatment. BV treatment temporarily reduced the relative abundance of BV-associated anaerobes (particularly Gardnerella vaginalis and Atopobium vaginae) and increased lactobacilli species (mainly L. iners), resulting in significantly altered mucosal immune milieu over time. In a linear mixed model, the median concentrations of pro-inflammatory cytokines and chemokines were significantly reduced in women who cleared BV compared to pre-treatment. BV persistence and recurrence were strongly associated with mucosal cytokine profiles that may increase the risk of HIV acquisition. Concentrations of these cytokines were differentially regulated by changes in the relative abundance of BVAB1 and G. vaginalis. We conclude that metronidazole for the treatment of BV induced short-term shifts in the vaginal microbiota and mucosal cytokines, while treatment failures promoted persistent elevation of pro-inflammatory cytokine concentrations in the genital tract. These data suggest the need to improve clinical management of BV to minimize BV related reproductive risk factors.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Bacteria/drug effects , Cytokines/metabolism , Inflammation Mediators/metabolism , Metronidazole/administration & dosage , Mucous Membrane/drug effects , Vagina/drug effects , Vaginosis, Bacterial/drug therapy , Administration, Oral , Adult , Anti-Bacterial Agents/adverse effects , Bacteria/immunology , Bacteria/pathogenicity , Dysbiosis , Female , Host-Pathogen Interactions , Humans , Longitudinal Studies , Metronidazole/adverse effects , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Prospective Studies , Reinfection , South Africa , Time Factors , Treatment Outcome , Vagina/immunology , Vagina/metabolism , Vagina/microbiology , Vaginosis, Bacterial/diagnosis , Vaginosis, Bacterial/immunology , Vaginosis, Bacterial/microbiology , Young Adult
13.
Front Immunol ; 12: 676669, 2021.
Article in English | MEDLINE | ID: mdl-34616391

ABSTRACT

Even with sustained use of antiretroviral therapy (ART), HIV-infected individuals have an increased risk of systemic comorbid conditions and oral pathologies, including opportunistic infections, oral mucosal inflammation, and gingival and periodontal diseases. The immune-mediated mechanisms that drive this increased risk, in the context of sustained viral suppression, are unclear. HIV infection, even when controlled, alters microbial communities contributing to a chronic low-grade inflammatory state that underlies these non-HIV co-morbidities. The higher prevalence of dental caries, and mucosal and periodontal inflammation reported in HIV-infected individuals on ART is often associated with differentially abundant oral microbial communities, possibly leading to a heightened susceptibility to inflammation. This mini-review highlights current gaps in knowledge regarding the microbe-mediated oral mucosal immunity with HIV infection while discussing opportunities for future research investigations and implementation of novel approaches to elucidate these gaps. Interventions targeting both inflammation and microbial diversity are needed to mitigate oral inflammation-related comorbidities, particularly in HIV-infected individuals. More broadly, additional research is needed to bolster general models of microbiome-mediated chronic immune activation and aid the development of precise microbiota-targeted interventions to reverse or mitigate adverse outcomes.


Subject(s)
HIV Infections/immunology , Immunity, Mucosal , Microbiota , Mucous Membrane/immunology , Mucous Membrane/microbiology , Anti-HIV Agents/therapeutic use , Dental Caries/complications , HIV Infections/drug therapy , HIV Infections/virology , Humans
14.
J Microbiol Methods ; 191: 106351, 2021 12.
Article in English | MEDLINE | ID: mdl-34710513

ABSTRACT

Human gut surface-attached mucosal microbiota plays significant roles in human health and diseases. This study sought to simulate the mucosal environment using mucin-agar gel and synthetic mucosal microbial community in vitro. To select suitable culture media, microbial communities were assembled and cultured in seven different media at 37 °C for 36 h. Among the seven media, Bryant & Burkey (BB) and Gifu Anaerobic Media (GAM) were selected considering their microbial biomass and bacterial composition. The communities were again assembled and cultured in these two media with mucin-agar. The results showed that some bacterial genus such as Bifidobacterium, Collinsella, and Roseburia could efficiently colonize in the solid mucin-agar part while Enterococcus, Clostridium, and Veilonella dominated in the liquid part. Metabolic functional prediction for the microbial community in each medium part showed that the gene expression involved in metabolism and cell motility pathways were distinctively differentiated between the liquid and solid medium part, and the functional potential was highly related to the microbial composition. The current results demonstrate that the simulation of the gut microbial ecosystem in vitro can be beneficial to the mucosal environment mimicking and the study on the mechanistic potential of the human gut microbiota for easy translation of microbiome research to therapies.


Subject(s)
Bacteriological Techniques/methods , Computer Simulation , Ecosystem , Gastrointestinal Microbiome , Mucous Membrane/microbiology , Agar , Biomass , Culture Media/chemistry , Diagnostic Tests, Routine , Enterococcus , Gastrointestinal Microbiome/genetics , Gene Expression , Genetic Techniques , Humans , Microbiota , Mucins
15.
BMC Microbiol ; 21(1): 244, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34488629

ABSTRACT

BACKGROUND: Fish skin represents an ancient vertebrate mucosal surface, sharing characteristics with other mucosal surfaces including those of the intestine. The skin mucosa is continuously exposed to microbes in the surrounding water and is therefore important in the first line defense against environmental pathogens by preventing bacteria from accessing the underlying surfaces. Understanding the microbe-host interactions at the fish skin mucosa is highly relevant in order to understand and control infection, commensalism, colonization, persistence, infection, and disease. Here we investigate the interactions between the pathogenic bacteria Aeromonas salmonicida (A. salmonicida) and Yersinia ruckeri (Y. ruckeri), respectively, and the skin mucosal surface of Atlantic salmon fry using AFM force spectroscopy. RESULTS: The results obtained revealed that when retracting probes functionalized with bacteria from surfaces coated with immobilized mucins, isolated from salmon mucosal surfaces, rupture events reflecting the disruption of adhesive interactions were observed, with rupture strengths centered around 200 pN. However, when retracting probes functionalized with bacteria from the intact mucosal surface of salmon fish fry no adhesive interactions could be detected. Furthermore, rheological measurements revealed a near fluid-like behavior for the fish fry skin mucus. Taken together, the experimental data indicate that the adhesion between the mucin molecules within the mucous layer may be significantly weaker than the interaction between the bacteria and the mucin molecules. The bacteria, immobilized on the AFM probe, do bind to individual mucins in the mucosal layer, but are released from the near fluid mucus with little resistance upon retraction of the AFM probe, to which they are immobilized. CONCLUSION: The data provided in the current paper reveal that A. salmonicida and Y. ruckeri do bind to the immobilized mucins. However, when retracting the bacteria from intact mucosal surfaces, no adhesive interactions are detected. These observations suggest a mechanism underlying the protective function of the mucosal surface based on the clearing of potential threats by adhering them to loosely attached mucus that is subsequently released from the fish skin.


Subject(s)
Bacterial Adhesion , Microscopy, Atomic Force/methods , Mucous Membrane/microbiology , Mucus/microbiology , Salmon/microbiology , Skin/microbiology , Aeromonas salmonicida/pathogenicity , Aeromonas salmonicida/physiology , Animals , Bacteria/classification , Bacteria/pathogenicity , Fish Diseases/microbiology , Mucus/metabolism , Yersinia ruckeri/pathogenicity , Yersinia ruckeri/physiology
16.
World J Microbiol Biotechnol ; 37(10): 176, 2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34519941

ABSTRACT

All mucins are highly glycosylated and a key constituent of the mucus layer that is vigilant against pathogens in many organ systems of animals and humans. The viscous layer is organized in bilayers, i.e., an outer layer that is loosely arranged, variable in thickness, home to the commensal microbiota that grows in the complex environment, and an innermost layer that is stratified, non-aspirated, firmly adherent to the epithelial cells and devoid of any microorganisms. The O-glycosylation moiety represents the site of adhesion for pathogens and due to the increase of motility, mucolytic activity, and upregulation of virulence factors, some microorganisms can circumvent the component of the mucus layer and cause disruption in organ homeostasis. A dysbiotic microbiome, defective mucus barrier, and altered immune response often result in various diseases. In this review, paramount emphasis is given to the role played by the bacterial species directly or indirectly involved in mucin degradation, alteration in mucus secretion or its composition or mucin gene expression, which instigates many diseases in the digestive, respiratory, and other organ systems. A systematic view can help better understand the etiology of some complex disorders such as cystic fibrosis, ulcerative colitis and expand our knowledge about mucin degraders to develop new therapeutic approaches to correct ill effects caused by these mucin-dwelling pathogens.


Subject(s)
Bacteria/metabolism , Bacterial Infections/microbiology , Mucous Membrane/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Infections/metabolism , Dysbiosis/metabolism , Dysbiosis/microbiology , Humans , Microbiota , Mucins/metabolism
17.
J Immunol ; 207(7): 1719-1724, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34544814

ABSTRACT

Metabolic diseases are common worldwide and include diseases of overnutrition, such as obesity, or undernutrition, such as kwashiorkor. Both the immune system and the microbiota contribute to a variety of metabolic diseases; however, these two processes have largely been studied independently of one another in this context. The gastrointestinal system houses the greatest density of microbes but also houses one of the largest collections of immune molecules, especially Abs. The IgA isotype dominates the Ab landscape at mucosal sites, and a number of studies have demonstrated the importance of this Ab to the stability of the microbiota. In this article, we review the literature that demonstrates how homeostatic Ab responses control microbiota composition and function to influence metabolic disease. We propose that many metabolic diseases may arise from disruptions to homeostatic immune control of gut commensals and that further understanding this interaction can offer a novel opportunity for therapeutic interventions.


Subject(s)
Dysbiosis/immunology , Immunoglobulin A/metabolism , Metabolic Diseases/immunology , Microbiota/immunology , Mucous Membrane/immunology , Animals , Dysbiosis/microbiology , Host Microbial Interactions , Humans , Immunity, Mucosal , Immunomodulation , Metabolic Diseases/microbiology , Mucous Membrane/microbiology
18.
Carbohydr Polym ; 273: 118595, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34560996

ABSTRACT

The microbial glycans mediate many significant biological acts, such as pathogen survival, host-microbe interactions, and immune evasion. The systematic study of microbial glycans structure remains challenging because of its high complexity and variability. In this study, we screened all the microbial glycans structures in the CSDB (Carbohydrate Structure Database), disassembled them into substructures, and calculated all the substructures' numbers. The results showed that a large number of glycan substructures are shared among different microorganisms. Further analysis showed that the glycan substructures appeared in specific bacterial groups may be related to the species and pathogenicity of microorganisms. Broadly, these findings provided an alternative approach or clue to discover the hidden information and the biological functions of glycans. The results can be used to detect broad-scope pathogen or prepare broad-spectrum vaccines.


Subject(s)
Bacteria/chemistry , Polysaccharides, Bacterial/chemistry , Polysaccharides/chemistry , Bacteria/classification , Bacteria/pathogenicity , Carbohydrate Sequence , Databases as Topic , Foodborne Diseases/microbiology , Fungal Polysaccharides/chemistry , Fungi/chemistry , Intestines/microbiology , Mucous Membrane/microbiology , Plants/microbiology , Wound Infection/microbiology
19.
Front Immunol ; 12: 726984, 2021.
Article in English | MEDLINE | ID: mdl-34421929

ABSTRACT

The moment a very old bacterial pathogen met a young virus from the 80's defined the beginning of a tragic syndemic for humanity. Such is the case for the causative agent of tuberculosis and the human immunodeficiency virus (HIV). Syndemic is by definition a convergence of more than one disease resulting in magnification of their burden. Both pathogens work synergistically contributing to speed up the replication of each other. Mycobacterium tuberculosis (Mtb) and HIV infections are in the 21st century among the leaders of morbidity and mortality of humankind. There is an urgent need for development of new approaches for prevention, better diagnosis, and new therapies for both infections. Moreover, these approaches should consider Mtb and HIV as a co-infection, rather than just as separate problems, to prevent further aggravation of the HIV-TB syndemic. Both pathogens manipulate the host immune responses to establish chronic infections in intracellular niches of their host cells. This includes manipulation of host relevant antimicrobial proteases such as cathepsins or their endogenous inhibitors. Here we discuss recent understanding on how Mtb and HIV interact with cathepsins and their inhibitors in their multifactorial functions during the pathogenesis of both infections. Particularly we will address the role on pathogen transmission, during establishment of intracellular chronic niches and in granuloma clinical outcome and tuberculosis diagnosis. This area of research will open new avenues for the design of innovative therapies and diagnostic interventions so urgently needed to fight this threat to humanity.


Subject(s)
Cathepsins/immunology , HIV Infections/immunology , Tuberculosis/immunology , Animals , Cathepsins/antagonists & inhibitors , Granuloma/immunology , HIV Infections/diagnosis , HIV Infections/transmission , Host-Pathogen Interactions , Humans , Immunity, Mucosal , Mucous Membrane/immunology , Mucous Membrane/microbiology , Tuberculosis/diagnosis , Tuberculosis/transmission
20.
Int J Mol Sci ; 22(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204294

ABSTRACT

Persistent infection with High Risk-Human Papilloma Viruses (HR-HPVs) is a primary cause of cervical cancer worldwide. Vaginal-dysbiosis-associated bacteria were correlated with the persistence of HR-HPVs infection and with increased cancer risk. We obtained strains of the most represented bacterial species in vaginal microbiota and evaluated their effects on the survival of cervical epithelial cells and immune homeostasis. The contribution of each species to supporting the antiviral response was also studied. Epithelial cell viability was affected by culture supernatants of most vaginal-dysbiosis bacteria, whereas Lactobacillus gasseri or Lactobacillus jensenii resulted in the best stimulus to induce interferon-γ (IFN-γ) production by human mononuclear cells from peripheral blood (PBMCs). Although vaginal-dysbiosis-associated bacteria induced the IFN-γ production, they were also optimal stimuli to interleukin-17 (IL-17) production. A positive correlation between IL-17 and IFN-γ secretion was observed in cultures of PBMCs with all vaginal-dysbiosis-associated bacteria suggesting that the adaptive immune response induced by these strains is not dominated by TH1 differentiation with reduced availability of IFN-γ, cytokine most effective in supporting virus clearance. Based on these results, we suggest that a vaginal microbiota dominated by lactobacilli, especially by L. gasseri or L. jensenii, may be able to assist immune cells with clearing HPV infection, bypasses the viral escape and restores immune homeostasis.


Subject(s)
Antibiosis , Dysbiosis , Homeostasis , Lactobacillus/physiology , Mucous Membrane/immunology , Mucous Membrane/microbiology , Vagina/immunology , Vagina/microbiology , Cell Survival , Cytokines/biosynthesis , Epithelial Cells/metabolism , Fatty Acids, Volatile/biosynthesis , Female , Humans , Vagina/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...