Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 678
Filtrar
2.
Biophys J ; 123(13): 1838-1845, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38824388

RESUMO

The gastrointestinal tract is inhabited by a vast community of microorganisms, termed the gut microbiota. Large colonies can pose a health threat, but the gastrointestinal mucus system protects epithelial cells from microbiota invasion. The human colon features a bilayer of mucus lining. Due to imbalances in intestinal homeostasis, bacteria may successfully penetrate the inner mucus layer, which can lead to severe gut diseases. However, it is hard to tease apart the competing mechanisms that lead to this penetration. To probe the conditions that permit bacteria penetration into the inner mucus layer, we develop an agent-based model consisting of bacteria and an inner mucus layer subject to a constant flux of nutrient fields feeding the bacteria. We find that there are three important variables that determine bacterial invasion: the bacterial reproduction rate, the contact energy between bacteria and mucus, and the rate of bacteria degrading the mucus. Under healthy conditions, all bacteria are naturally eliminated by the constant removal of mucus. In diseased states, imbalances between the rates of bacterial degradation and mucus secretion lead to bacterial invasion at certain junctures. We conduct uncertainty quantification and sensitivity analysis to compare the relative impact between these parameters. The contact energy has the strongest influence on bacterial penetration, which, in combination with bacterial degradation rate and growth rate, greatly accelerates bacterial invasion of the human gut mucus lining. Our findings will serve as predictive indicators for the etiology of intestinal diseases and highlight important considerations when developing gut therapeutics.


Assuntos
Colo , Modelos Biológicos , Muco , Humanos , Colo/microbiologia , Muco/metabolismo , Muco/microbiologia , Bactérias/metabolismo , Microbioma Gastrointestinal
3.
mSphere ; 9(6): e0008124, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38837404

RESUMO

In a healthy colon, the stratified mucus layer serves as a crucial innate immune barrier to protect the epithelium from microbes. Mucins are complex glycoproteins that serve as a nutrient source for resident microflora and can be exploited by pathogens. We aimed to understand how the intestinal pathogen, Clostridioides difficile, independently uses or manipulates mucus to its benefit, without contributions from members of the microbiota. Using a 2-D primary human intestinal epithelial cell model to generate physiologic mucus, we assessed C. difficile-mucus interactions through growth assays, RNA-Seq, biophysical characterization of mucus, and contextualized metabolic modeling. We found that host-derived mucus promotes C. difficile growth both in vitro and in an infection model. RNA-Seq revealed significant upregulation of genes related to central metabolism in response to mucus, including genes involved in sugar uptake, the Wood-Ljungdahl pathway, and the glycine cleavage system. In addition, we identified differential expression of genes related to sensing and transcriptional control. Analysis of mutants with deletions in highly upregulated genes reflected the complexity of C. difficile-mucus interactions, with potential interplay between sensing and growth. Mucus also stimulated biofilm formation in vitro, which may in turn alter the viscoelastic properties of mucus. Context-specific metabolic modeling confirmed differential metabolism and the predicted importance of enzymes related to serine and glycine catabolism with mucus. Subsequent growth experiments supported these findings, indicating mucus is an important source of serine. Our results better define responses of C. difficile to human gastrointestinal mucus and highlight flexibility in metabolism that may influence pathogenesis. IMPORTANCE: Clostridioides difficile results in upward of 250,000 infections and 12,000 deaths annually in the United States. Community-acquired infections continue to rise, and recurrent disease is common, emphasizing a vital need to understand C. difficile pathogenesis. C. difficile undoubtedly interacts with colonic mucus, but the extent to which the pathogen can independently respond to and take advantage of this niche has not been explored extensively. Moreover, the metabolic complexity of C. difficile remains poorly understood but likely impacts its capacity to grow and persist in the host. Here, we demonstrate that C. difficile uses native colonic mucus for growth, indicating C. difficile possesses mechanisms to exploit the mucosal niche. Furthermore, mucus induces metabolic shifts and biofilm formation in C. difficile, which has potential ramifications for intestinal colonization. Overall, our work is crucial to better understand the dynamics of C. difficile-mucus interactions in the context of the human gut.


Assuntos
Biofilmes , Clostridioides difficile , Regulação Bacteriana da Expressão Gênica , Muco , Clostridioides difficile/genética , Clostridioides difficile/fisiologia , Clostridioides difficile/metabolismo , Biofilmes/crescimento & desenvolvimento , Humanos , Muco/microbiologia , Muco/metabolismo , Células Epiteliais/microbiologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , Infecções por Clostridium/microbiologia
4.
J Agric Food Chem ; 72(27): 15345-15356, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38932522

RESUMO

The human intestinal mucus layer protects against pathogenic microorganisms and harmful substances, whereas it also provides an important colonization niche for mutualistic microbes. The main functional components of mucus are heavily glycosylated proteins, called mucins. Mucins can be cleaved and utilized by intestinal microbes. The mechanisms between intestinal microbes and the regulation of mucin glycosylation are still poorly understood. In this study, in vitro mucus was produced by HT29-MTX-E12 cells under Semi-Wet interface with Mechanical Stimulation. Cells were exposed to pasteurized nonpathogenic bacteria Akkermansia muciniphila, Ruminococcus gnavus, and Bacteroides fragilis to evaluate influence on glycosylation patterns. Following an optimized protocol, O- and N-glycans were efficiently and reproducibly released, identified, and semiquantified using MALDI-TOF-MS and PGC-LC-MS/MS. Exposure of cells to bacteria demonstrated increased diversity of sialylated O-glycans and increased abundance of high mannose N-glycans in in vitro produced mucus. Furthermore, changes in glycan ratios were observed. It is speculated that bacterial components interact with the enzymatic processes in glycan production and that pasteurized bacteria influence glycosyltransferases or genes involved. These results highlight the influence of pasteurized bacteria on glycosylation patterns, stress the intrinsic relationship between glycosylation and microbiota, and show the potential of using in vitro produced mucus to study glycosylation behavior.


Assuntos
Microbioma Gastrointestinal , Muco , Polissacarídeos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem , Glicosilação , Humanos , Espectrometria de Massas em Tandem/métodos , Muco/microbiologia , Muco/metabolismo , Muco/química , Polissacarídeos/metabolismo , Polissacarídeos/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Mucinas/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Bactérias/metabolismo , Bactérias/classificação , Bactérias/genética , Células HT29 , Cromatografia Líquida/métodos , Bacteroides fragilis/metabolismo , Bacteroides fragilis/química , Bacteroides fragilis/fisiologia , Pasteurização , Akkermansia/metabolismo , Espectrometria de Massa com Cromatografia Líquida
5.
PeerJ ; 12: e17421, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38827308

RESUMO

Background: Rainfall-induced coastal runoff represents an important environmental impact in near-shore coral reefs that may affect coral-associated bacterial microbiomes. Shifts in microbiome community composition and function can stress corals and ultimately cause mortality and reef declines. Impacts of environmental stress may be site specific and differ between coral microbiome compartments (e.g., tissue versus mucus). Coastal runoff and associated water pollution represent a major stressor for near-shore reef-ecosystems in Guam, Micronesia. Methods: Acropora pulchra colonies growing on the West Hagåtña reef flat in Guam were sampled over a period of 8 months spanning the 2021 wet and dry seasons. To examine bacterial microbiome diversity and composition, samples of A. pulchra tissue and mucus were collected during late April, early July, late September, and at the end of December. Samples were collected from populations in two different habitat zones, near the reef crest (farshore) and close to shore (nearshore). Seawater samples were collected during the same time period to evaluate microbiome dynamics of the waters surrounding coral colonies. Tissue, mucus, and seawater microbiomes were characterized using 16S DNA metabarcoding in conjunction with Illumina sequencing. In addition, water samples were collected to determine fecal indicator bacteria (FIB) concentrations as an indicator of water pollution. Water temperatures were recorded using data loggers and precipitation data obtained from a nearby rain gauge. The correlation structure of environmental parameters (temperature and rainfall), FIB concentrations, and A. pulchra microbiome diversity was evaluated using a structural equation model. Beta diversity analyses were used to investigate spatio-temporal trends of microbiome composition. Results: Acropora pulchra microbiome diversity differed between tissues and mucus, with mucus microbiome diversity being similar to the surrounding seawater. Rainfall and associated fluctuations of FIB concentrations were correlated with changes in tissue and mucus microbiomes, indicating their role as drivers of A. pulchra microbiome diversity. A. pulchra tissue microbiome composition remained relatively stable throughout dry and wet seasons; tissues were dominated by Endozoicomonadaceae, coral endosymbionts and putative indicators of coral health. In nearshore A. pulchra tissue microbiomes, Simkaniaceae, putative obligate coral endosymbionts, were more abundant than in A. pulchra colonies growing near the reef crest (farshore). A. pulchra mucus microbiomes were more diverse during the wet season than the dry season, a distinction that was also associated with drastic shifts in microbiome composition. This study highlights the seasonal dynamics of coral microbiomes and demonstrates that microbiome diversity and composition may differ between coral tissues and the surface mucus layer.


Assuntos
Antozoários , Recifes de Corais , Microbiota , Estações do Ano , Animais , Antozoários/microbiologia , Microbiota/fisiologia , Microbiota/genética , Muco/microbiologia , Água do Mar/microbiologia , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação
6.
Nat Commun ; 15(1): 4578, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38811586

RESUMO

Modulation of the cervix by steroid hormones and commensal microbiome play a central role in the health of the female reproductive tract. Here we describe organ-on-a-chip (Organ Chip) models that recreate the human cervical epithelial-stromal interface with a functional epithelial barrier and production of mucus with biochemical and hormone-responsive properties similar to living cervix. When Cervix Chips are populated with optimal healthy versus dysbiotic microbial communities (dominated by Lactobacillus crispatus and Gardnerella vaginalis, respectively), significant differences in tissue innate immune responses, barrier function, cell viability, proteome, and mucus composition are observed that are similar to those seen in vivo. Thus, human Cervix Organ Chips represent physiologically relevant in vitro models to study cervix physiology and host-microbiome interactions, and hence may be used as a preclinical testbed for development of therapeutic interventions to enhance women's health.


Assuntos
Colo do Útero , Interações entre Hospedeiro e Microrganismos , Imunidade Inata , Microbiota , Humanos , Feminino , Colo do Útero/microbiologia , Colo do Útero/imunologia , Microbiota/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Gardnerella vaginalis/imunologia , Lactobacillus crispatus/imunologia , Muco/imunologia , Muco/microbiologia , Muco/metabolismo , Dispositivos Lab-On-A-Chip
7.
mBio ; 15(6): e0345123, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38651896

RESUMO

The airway milieu of individuals with muco-obstructive airway diseases (MADs) is defined by the accumulation of dehydrated mucus due to hyperabsorption of airway surface liquid and defective mucociliary clearance. Pathological mucus becomes progressively more viscous with age and disease severity due to the concentration and overproduction of mucin and accumulation of host-derived extracellular DNA (eDNA). Respiratory mucus of MADs provides a niche for recurrent and persistent colonization by respiratory pathogens, including Pseudomonas aeruginosa, which is responsible for the majority of morbidity and mortality in MADs. Despite high concentration inhaled antibiotic therapies and the absence of antibiotic resistance, antipseudomonal treatment failure in MADs remains a significant clinical challenge. Understanding the drivers of antibiotic tolerance is essential for developing more effective treatments that eradicate persistent infections. The complex and dynamic environment of diseased airways makes it difficult to model antibiotic efficacy in vitro. We aimed to understand how mucin and eDNA concentrations, the two dominant polymers in respiratory mucus, alter the antibiotic tolerance of P. aeruginosa. Our results demonstrate that polymer concentration and molecular weight affect P. aeruginosa survival post antibiotic challenge. Polymer-driven antibiotic tolerance was not explicitly associated with reduced antibiotic diffusion. Lastly, we established a robust and standardized in vitro model for recapitulating the ex vivo antibiotic tolerance of P. aeruginosa observed in expectorated sputum across age, underlying MAD etiology, and disease severity, which revealed the inherent variability in intrinsic antibiotic tolerance of host-evolved P. aeruginosa populations. IMPORTANCE: Antibiotic treatment failure in Pseudomonas aeruginosa chronic lung infections is associated with increased morbidity and mortality, illustrating the clinical challenge of bacterial infection control. Understanding the underlying infection environment, as well as the host and bacterial factors driving antibiotic tolerance and the ability to accurately recapitulate these factors in vitro, is crucial for improving antibiotic treatment outcomes. Here, we demonstrate that increasing concentration and molecular weight of mucin and host eDNA drive increased antibiotic tolerance to tobramycin. Through systematic testing and modeling, we identified a biologically relevant in vitro condition that recapitulates antibiotic tolerance observed in ex vivo treated sputum. Ultimately, this study revealed a dominant effect of in vivo evolved bacterial populations in defining inter-subject ex vivo antibiotic tolerance and establishes a robust and translatable in vitro model for therapeutic development.


Assuntos
Antibacterianos , Muco , Infecções por Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Muco/microbiologia , Muco/metabolismo , Humanos , Mucinas/metabolismo , Farmacorresistência Bacteriana , Polímeros/metabolismo , Infecção Persistente/microbiologia , Pulmão/microbiologia , Infecções Respiratórias/microbiologia , Infecções Respiratórias/tratamento farmacológico , Adaptação Fisiológica
8.
Fish Shellfish Immunol ; 149: 109527, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38561068

RESUMO

Skin mucus analysis has recently been used as a non-invasive method to evaluate for fish welfare. The present research study was conducted to examine the skin mucosal immunity and skin microbiota profiles of sturgeons infected with Citrobacter freundii. Our histology results showed that the thickness of the epidermal layer of skin remained thinner, and the number of mucous cells was significantly decreased in sturgeons after infection (p < 0.05). Total protein, alanine aminotransferase, aspartate aminotransferase, superoxide dismutase, and creatine kinase levels in the mucus showed biphasic pattern (decrease and then increase). Lactate dehydrogenase, lysozyme, and acid phosphatase activities in the mucus showed an increasing trend after infection. Furthermore, 16S rRNA sequencing also revealed that C. freundii infection also affected the diversity and community structure of the skin mucus microbiota. An increase in microbial diversity (p > 0.05) and a decrease in microbial abundance (p < 0.05) after infection were noted. The predominant bacterial phyla in the skin mucus were Proteobacteria, Fusobacteria, Bacteroidetes, Firmicutes, and Actinobacteria. Specifically, the relative abundance of Fusobacteria increased after infection. The predominant bacterial genera in the skin mucus were Cetobacterium, Pelomonas, Bradyrhizobium, Flavobacterium, and Pseudomonas. The relative abundance of Cetobacterium, Pseudomonas, and Flavobacterium increased after infection. Our current research findings will provide new insights into the theoretical basis for future research studies exploring the mechanism of sturgeon infection with C. freundii.


Assuntos
Citrobacter freundii , Infecções por Enterobacteriaceae , Doenças dos Peixes , Peixes , Imunidade nas Mucosas , Microbiota , Pele , Animais , Citrobacter freundii/imunologia , Microbiota/imunologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Pele/imunologia , Pele/microbiologia , Peixes/imunologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/veterinária , Infecções por Enterobacteriaceae/microbiologia , Muco/imunologia , Muco/microbiologia , RNA Ribossômico 16S/genética
9.
An Acad Bras Cienc ; 95(4): e20200325, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38055606

RESUMO

Mucus, produced by Palythoa caribaeorum has been popularly reported due to healing, anti-inflammatory, and analgesic effects. However, biochemical and pharmacological properties of this mucus remains unexplored. Therefore, the present study aimed to study its proteome profile by 2DE electrophoresis and MALDI-TOF. Furthermore, it was evaluated the cytotoxic, antibacterial, and antioxidant activities of the mucus and from its protein extract (PE). Proteomics study identified14 proteins including proteins involved in the process of tissue regeneration and death of tumor cells. The PE exhibited cell viability below 50% in the MCF-7 and S-180 strains. It showed IC50 of 6.9 µg/mL for the J774 lineage, and also, favored the cellular growth of fibroblasts. Furthermore, PE revealed activity against Escherichia coli, Klebsiella pneumoniae, Staphylococcus aureus, and Staphylococcus epidermidis (MIC of 250 µg/mL). These findings revealed the mucus produced by Palythoa caribaeorum with biological activities, offering alternative therapies for the treatment of cancer and as a potential antibacterial agent.


Assuntos
Antozoários , Proteômica , Animais , Antozoários/química , Antibacterianos/farmacologia , Proteínas , Muco/microbiologia , Testes de Sensibilidade Microbiana
10.
Microbiol Spectr ; 11(4): e0033623, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37289064

RESUMO

Culturing the gut microbiota in in vitro models that mimic the intestinal environment is increasingly becoming a promising alternative approach to study microbial dynamics and the effect of perturbations on the gut community. Since the mucus-associated microbial populations in the human intestine differ in composition and functions from their luminal counterpart, we attempted to reproduce in vitro the microbial consortia adhering to mucus using an already established three-dimensional model of the human gut microbiota. Electrospun gelatin structures supplemented or not with mucins were inoculated with fecal samples and compared for their ability to support microbial adhesion and growth over time, as well as to shape the composition of the colonizing communities. Both scaffolds allowed the establishment of long-term stable biofilms with comparable total bacterial loads and biodiversity. However, mucin-coated structures harbored microbial consortia especially enriched in Akkermansia, Lactobacillus, and Faecalibacterium, being therefore able to select for microorganisms commonly considered mucosa-associated in vivo. IMPORTANCE These findings highlight the importance of mucins in shaping intestinal microbial communities, even those in artificial gut microbiota systems. We propose our in vitro model based on mucin-coated electrospun gelatin structures as a valid device for studies evaluating the effects of exogenous factors (nutrients, probiotics, infectious agents, and drugs) on mucus-adhering microbial communities.


Assuntos
Microbioma Gastrointestinal , Humanos , Gelatina/farmacologia , Bactérias , Mucinas/química , Mucinas/farmacologia , Muco/microbiologia , Mucosa Intestinal/microbiologia
11.
ACS Biomater Sci Eng ; 9(6): 2819-2837, 2023 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-34784462

RESUMO

The gastrointestinal mucus layer plays a significant role in maintaining gut homeostasis and health, offering protective capacities against the absorption of harmful pathogens as well as commensal gut bacteria and buffering stomach acid to protect the underlying epithelium. Despite this, the mucus barrier is often overlooked during preclinical pharmaceutical development and may pose a significant absorption barrier to high molecular weight or lipophilic drug species. The complex chemical and physical nature of the dynamic mucus layer has proven problematic to reliably replicate in a laboratory setting, leading to the development of multiple mucus models with varying complexity and predictive capacity. This, coupled with the wide range of analysis methods available, has led to a plethora of possible approaches to quantifying mucus permeation; however, the field remains significantly under-represented in biomedical research. For this reason, the development of a concise collation of the available approaches to mucus permeation is essential. In this review, we explore widely utilized mucus mimics ranging in complexity from simple mucin solutions to native mucus preparations for their predictive capacity in mucus permeation analysis. Furthermore, we highlight the diverse range of laboratory-based models available for the analysis of mucus interaction and permeability with a specific focus on in vitro, ex vivo, and in situ models. Finally, we highlight the predictive capacity of these models in correlation with in vivo pharmacokinetic data. This review provides a comprehensive and critical overview of the available technologies to analyze mucus permeation, facilitating the efficient selection of appropriate tools for further advancement in oral drug delivery.


Assuntos
Sistemas de Liberação de Medicamentos , Muco , Preparações Farmacêuticas/análise , Preparações Farmacêuticas/química , Permeabilidade , Muco/química , Muco/microbiologia , Bactérias
12.
Sci Rep ; 12(1): 18193, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36307456

RESUMO

The outer mucus layer of the colorectal epithelium is easily removable and colonized by commensal microbiota, while the inner mucus layer is firmly attached to the epithelium and devoid of bacteria. Although the specific bacteria penetrating the inner mucus layer can contact epithelial cells and trigger cancer development, most studies ignore the degree of mucus adhesion at sampling. Therefore, we evaluated whether bacteria adhering to tissues could be identified by removing the outer mucus layer. Our 16S rRNA gene sequencing analysis of 18 surgical specimens of human colorectal cancer revealed that Sutterella (P = 0.045) and Enterobacteriaceae (P = 0.045) were significantly enriched in the mucus covering the mucosa relative to the mucosa. Rikenellaceae (P = 0.026) was significantly enriched in the mucus covering cancer tissues compared with those same cancer tissues. Ruminococcaceae (P = 0.015), Enterobacteriaceae (P = 0.030), and Erysipelotrichaceae (P = 0.028) were significantly enriched in the mucus covering the mucosa compared with the mucus covering cancers. Fusobacterium (P = 0.038) was significantly enriched in the mucus covering cancers compared with the mucus covering the mucosa. Comparing the microbiomes of mucus and tissues with mucus removed may facilitate identifying bacteria that genuinely invade tissues and affect tumorigenesis.


Assuntos
Neoplasias Colorretais , Microbiota , Humanos , Colo/microbiologia , Mucosa Intestinal/microbiologia , RNA Ribossômico 16S/genética , Muco/microbiologia , Microbiota/genética , Bactérias/genética , Neoplasias Colorretais/genética
13.
World J Gastroenterol ; 28(32): 4475-4492, 2022 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-36157924

RESUMO

Little was known about mammalian colon mucus (CM) until the beginning of the 21st century. Since that time considerable progress has been made in basic research addressing CM structure and functions. Human CM is formed by two distinct layers composed of gel-forming glycosylated mucins that are permanently secreted by goblet cells of the colonic epithelium. The inner layer is dense and impenetrable for bacteria, whereas the loose outer layer provides a habitat for abundant commensal microbiota. Mucus barrier integrity is essential for preventing bacterial contact with the mucosal epithelium and maintaining homeostasis in the gut, but it can be impaired by a variety of factors, including CM-damaging switch of commensal bacteria to mucin glycan consumption due to dietary fiber deficiency. It is proven that impairments in CM structure and function can lead to colonic barrier deterioration that opens direct bacterial access to the epithelium. Bacteria-induced damage dysregulates epithelial proliferation and causes mucosal inflammatory responses that may expand to the loosened CM and eventually result in severe disorders, including colitis and neoplastic growth. Recently described formation of bacterial biofilms within the inner CM layer was shown to be associated with both inflammation and cancer. Although obvious gaps in our knowledge of human CM remain, its importance for the pathogenesis of major colorectal diseases, comprising inflammatory bowel disease and colorectal cancer, is already recognized. Continuing progress in CM exploration is likely to result in the development of a range of new useful clinical applications addressing colorectal disease diagnosis, prevention and therapy.


Assuntos
Neoplasias Colorretais , Muco , Animais , Colo/patologia , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/patologia , Fibras na Dieta , Humanos , Mucosa Intestinal/microbiologia , Mamíferos , Mucinas , Muco/microbiologia , Muco/fisiologia , Polissacarídeos
14.
Gut Microbes ; 14(1): 2041342, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35239459

RESUMO

The immune system in the large intestine is separated from commensal microbes and comparatively rare enteric pathogens by a monolayer of diverse epithelial cells overlaid with a compact and adherent inner mucus layer and a looser outer mucus layer. Microorganisms, collectively referred to as the mucus-associated (MA) microbiota, physically inhabit this mucus barrier, resulting in a dynamic and incessant dialog to maintain both spatial segregation and immune tolerance. Recent major findings reveal novel features of the crosstalk between the immune system and mucus-associated bacteria in health and disease, as well as disease-related peripheral immune signatures indicative of host responses to these organisms. In this brief review, we integrate these novel observations into our overall understanding of host-microbiota mutualism at the colonic mucosal border and speculate on the significance of this emerging knowledge for our understanding of the prevention, development, and progression of chronic intestinal inflammation.


Assuntos
Microbioma Gastrointestinal , Simbiose , Colo/microbiologia , Humanos , Sistema Imunitário , Inflamação , Mucosa Intestinal/microbiologia , Muco/microbiologia
15.
Digestion ; 103(3): 232-243, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35184056

RESUMO

BACKGROUND: The pathophysiology of ulcerative colitis (UC) remains unclear, but early lesions on the colorectal mucosal surface may play an important role in its etiology. Intestinal mucus samples, including inner and outer layers, are collected by net or brush catheters, but the quality of the samples obtained by each method has not been fully investigated. OBJECTIVE: The purpose of this study was to compare the microbiome and protein content of intestinal mucus collected by net and brush catheters during colonoscopy. METHODS: Intestinal mucus samples from the lower rectum of 4 patients with UC were collected using a net catheter, a brush catheter, and intestinal fluid suction. Microbiome and protein content were analyzed using 16S rRNA gene sequencing and mass spectrometry. RESULTS: The patients demonstrated significant differences in microbiome alpha diversity (p < 0.05), but this difference was not observed between the sampling methods. Net catheter samples demonstrated higher total protein concentrations than brush catheter samples. The brush catheter group had more Lachnospira, a butyrate-producing bacterium, when compared to the net group. The brush catheter group also had more oral bacteria of Staphylococcus and Dialister in those with active phase when compared to the net group. CONCLUSIONS: Brush catheters are more likely to collect the intestinal mucus inner layer, whereas net catheters are more likely to collect larger samples that include the outer mucus layer, as well as the intestinal fluid. Two sampling methods with different types of collection of the mucosa may lead to different results among patients with mucosal vulnerabilities.


Assuntos
Colite Ulcerativa , Catéteres , Colite Ulcerativa/patologia , Humanos , Mucosa Intestinal/metabolismo , Muco/metabolismo , Muco/microbiologia , RNA Ribossômico 16S/genética , Reto/patologia
16.
Food Funct ; 13(6): 3098-3109, 2022 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-35226005

RESUMO

As a broadly defined member of lactic acid bacteria (LAB), the Lactobacillus strain is well characterized in food fermentation and specific strains can enhance the intestinal barrier function and be recognized as the probiotic strain. In recent years, many molecules of the cell surface are thought to be related to the adhesion property in the gastrointestinal mucosa. Mucus layer-related proteins, extracellular matrix proteins, and immunoglobulins also exhibit immunity regulation and protection of the intestinal epithelial barrier function. Meanwhile, the effects of bile and the low pH of the gastrointestinal tract (GIT) on Lactobacillus colonization are also needed to be considered. Furthermore, LAB can adhere and aggregate in the GIT to promote the maturity of biofilm and the extracellular matrix secreting through the signal molecules in the quorum sensing (QS) system. Therefore, it is of great interest to use the QS system to regulate the initial adhesion ability of Lactobacillus and further enhance the probiotic effect of the biofilm formation of beneficial bacteria. This review summarizes the adhesion properties of cell surface proteins derived from Lactobacillus strains in recent studies and provides valuable information on the QS effect on the adhesion property of Lactobacillus strains in the GIT environment.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Trato Gastrointestinal/microbiologia , Lactobacillales/fisiologia , Lactobacillus/fisiologia , Proteínas de Membrana/metabolismo , Percepção de Quorum , Fímbrias Bacterianas/fisiologia , Flagelos/fisiologia , Humanos , Lactobacillus/ultraestrutura , Glicoproteínas de Membrana/metabolismo , Muco/metabolismo , Muco/microbiologia , Peptidoglicano/química , Peptidoglicano/metabolismo , Probióticos , Ácidos Teicoicos/química , Ácidos Teicoicos/metabolismo
17.
Microbiologyopen ; 11(1): e1263, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35212476

RESUMO

The search for new natural compounds for application in medicine and cosmetics is a trend in biotechnology. One of the sources of such active compounds is the snail mucus. Snail physiology and the biological activity of their fluids (especially the mucus) are still poorly studied. Only a few previous studies explored the relationship between snails and their microbiome. The present study was focused on the biodiversity of the snail mucus used in the creation of cosmetic products, therapeutics, and nutraceuticals. The commonly used cultivation techniques were applied for the determination of the number of major bacterial groups. Fluorescence in situ hybridization for key taxa was performed. The obtained images were subjected to digital image analysis. Sequencing of the 16S rRNA gene was also done. The results showed that the mucus harbors a rich bacterial community (10.78 × 1010 CFU/ml). Among the dominant bacteria, some are known for their ability to metabolize complex polysaccharides or are usually found in soil and plants (Rhizobiaceae, Shewanella, Pedobacter, Acinetobacter, Alcaligenes). The obtained data demonstrated that the snail mucus creates a unique environment for the development of the microbial community that differs from other parts of the animal and which resulted from the combined contribution of the microbiomes derived from the soil, plants, and the snails.


Assuntos
Bactérias/isolamento & purificação , Caramujos/microbiologia , Sequência de Aminoácidos , Animais , Bactérias/classificação , Bactérias/genética , Biologia Computacional , Hibridização in Situ Fluorescente , Ponto Isoelétrico , Metagenômica , Microbiota , Muco/química , Muco/microbiologia , RNA Ribossômico 16S/genética , Caramujos/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem
18.
Trends Mol Med ; 28(1): 36-50, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34810087

RESUMO

The prevalence of food allergies has reached epidemic levels but the cause remains largely unknown. We discuss the clinical relevance of the gut mucosal barrier as a site for allergic sensitization to food. In this context, we focus on an important but overlooked part of the mucosal barrier in pathogenesis, the glycoprotein-rich mucus layer, and call attention to both beneficial and detrimental aspects of mucus-gut microbiome interactions. Studying the intricate links between the mucus barrier, the associated bacteria, and the mucosal immune system may advance our understanding of the mechanisms and inform prevention and treatment strategies in food allergy.


Assuntos
Hipersensibilidade Alimentar , Microbioma Gastrointestinal , Bactérias , Hipersensibilidade Alimentar/epidemiologia , Hipersensibilidade Alimentar/terapia , Humanos , Mucosa Intestinal/microbiologia , Muco/microbiologia
19.
Angew Chem Int Ed Engl ; 61(3): e202112346, 2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-34816559

RESUMO

Detection of pathogenic bacteria in complex biological matrices remains a major challenge. Herein, we report the selection and optimization of a new DNAzyme for Staphylococcus aureus (SA) and the use of the DNAzyme to develop a simple lateral flow device (LFD) for detection of SA in nasal mucus. The DNAzyme was generated by in vitro selection using a crude extra/intracellular mixture derived from SA, which could be used directly for simple solution or paper-based fluorescence assays for SA. The DNAzyme was further modified to produce a DNA cleavage fragment that acted as a bridging element to bind DNA-modified gold nanoparticles to the test line of a LFD, producing a simple colorimetric dipstick test. The LFD was evaluated with nasal mucus samples spiked with SA, and demonstrated that SA detection was possible in minutes with minimal sample processing.


Assuntos
Técnicas Biossensoriais , DNA Catalítico/metabolismo , Muco/microbiologia , Cavidade Nasal/microbiologia , Staphylococcus aureus/isolamento & purificação , Humanos , Staphylococcus aureus/metabolismo
20.
Int J Syst Evol Microbiol ; 71(11)2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34797211

RESUMO

A study on the polyphasic taxonomic classification of an Arcobacter strain, R-73987T, isolated from the rectal mucus of a porcine intestinal tract, was performed. Phylogenetic analysis based on the 16S rRNA gene sequence revealed that the strain could be assigned to the genus Arcobacter and suggested that strain R-73987T belongs to a novel undescribed species. Comparative analysis of the rpoB gene sequence confirmed the findings. Arcobacter faecis LMG 28519T was identified as its closest neighbour in a multigene analysis based on 107 protein- encoding genes. Further, whole-genome sequence comparisons by means of average nucleotide identity and in silico DNA-DNA hybridization between the genome of strain R-73987T and the genomes of validly named Arcobacter species resulted in values below 95-96 and 70  %, respectively. In addition, a phenotypic analysis further corroborated the conclusion that strain R-73987T represents a novel Arcobacter species, for which the name Arcobacter vandammei sp. nov. is proposed. The type strain is R-73987T (=LMG 31429T=CCUG 75005T). This appears to be the first Arcobacter species recovered from porcine intestinal mucus.


Assuntos
Arcobacter , Filogenia , Reto/microbiologia , Sus scrofa/microbiologia , Animais , Arcobacter/classificação , Arcobacter/isolamento & purificação , Técnicas de Tipagem Bacteriana , Composição de Bases , DNA Bacteriano/genética , Ácidos Graxos/química , Muco/microbiologia , Hibridização de Ácido Nucleico , RNA Ribossômico 16S/genética , Análise de Sequência de DNA , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA