Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Gut Microbes ; 13(1): 1-23, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33789528

RESUMO

Human milk oligosaccharides (HMOs) and milk fat globule membrane (MFGM) are highly abundant in breast milk, and have been shown to exhibit potent immunomodulatory effects. Yet, their role in the gut microbiota modulation in relation to colitis remains understudied. Since the mixtures of fructo-oligosaccharides (FOS) and galacto-oligosaccharides (GOS) perfectly mimic the properties and functions of HMOs, the combination of MFGM, FOS, and GOS (CMFG) has therefore been developed and used in this study. Here, CMFG were pre-fed to mice for three weeks to investigate its preventive effect on dextran sodium sulfate (DSS) induced colitis. Moreover, CMFG-treated and vehicle-treated mice were cohoused to further elucidate the preventive role of the gut microbiota transfer in colitis. At the end of the study, 16S rDNA gene amplicon sequencing, short-chain fatty acids (SCFAs) profiling, transcriptome sequencing, histological analysis, immunofluorescence staining and flow cytometry analysis were conducted. Our results showed that CMFG pre-supplementation alleviated DSS-induced colitis as evidenced by decreased disease activity index (DAI) score, reduced body weight loss, increased colon length and mucin secretion, and ameliorated intestinal damage. Moreover, CMFG reduced macrophages in the colon, resulting in decreased levels of IL-1ß, IL-6, IL-8, TNF-α, and MPO in the colon and circulation. Furthermore, CMFG altered the gut microbiota composition and promoted SCFAs production in DSS-induced colitis. Markedly, the cohousing study revealed that transfer of gut microbiota from CMFG-treated mice largely improved the DSS-induced colitis as evidenced by reduced intestinal damage and decreased macrophages infiltration in the colon. Moreover, transfer of the gut microbiota from CMFG-treated mice protected against DSS-induced gut microbiota dysbiosis and promotes SCFAs production, which showed to be associated with colitis amelioration. Collectively, these findings demonstrate the beneficial role of CMFG in the gastrointestinal diseases, and further provide evidence for the rational design of effective prophylactic functional diets in both animals and humans.


Assuntos
Colite/tratamento farmacológico , Colo/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Glicolipídeos/farmacologia , Glicoproteínas/farmacologia , Homeostase , Macrófagos/metabolismo , Oligossacarídeos/farmacologia , Animais , Colite/induzido quimicamente , Colite/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Disbiose/microbiologia , Ácidos Graxos Voláteis/metabolismo , Feminino , Glicolipídeos/metabolismo , Glicoproteínas/metabolismo , Humanos , Gotículas Lipídicas/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Leite Humano , Mucosa/metabolismo , Muco/microbiologia , Oligossacarídeos/metabolismo , RNA Ribossômico 16S , Sequenciamento do Exoma
7.
Front Immunol ; 12: 633621, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777020

RESUMO

One of the main targets for the use of phytogenics in aquafeeds is the mucosal tissues as they constitute a physical and biochemical shield against environmental and pathogenic threats, comprising elements from both the innate and acquired immunity. In the present study, the modulation of the skin transcriptional immune response, the bacterial growth capacity in skin mucus, and the overall health condition of gilthead seabream (Sparus aurata) juveniles fed a dietary supplementation of garlic essential oil, carvacrol, and thymol were assessed. The enrichment analysis of the skin transcriptional profile of fish fed the phytogenic-supplemented diet revealed the regulation of genes associated to cellular components involved in the secretory pathway, suggesting the stimulation, and recruitment of phagocytic cells. Genes recognized by their involvement in non-specific immune response were also identified in the analysis. The promotion of the secretion of non-specific immune molecules into the skin mucus was proposed to be involved in the in vitro decreased growth capacity of pathogenic bacteria in the mucus of fish fed the phytogenic-supplemented diet. Although the mucus antioxidant capacity was not affected by the phytogenics supplementation, the regulation of genes coding for oxidative stress enzymes suggested the reduction of the skin oxidative stress. Additionally, the decreased levels of cortisol in mucus indicated a reduction in the fish allostatic load due to the properties of the tested additive. Altogether, the dietary garlic, carvacrol, and thymol appear to promote the gilthead seabream skin innate immunity and the mucus protective capacity, decreasing its susceptibility to be colonized by pathogenic bacteria.


Assuntos
Imunidade Inata/efeitos dos fármacos , Muco/metabolismo , Óleos Voláteis/farmacologia , Dourada/imunologia , Via Secretória/efeitos dos fármacos , Pele/efeitos dos fármacos , Ração Animal/análise , Animais , Aquicultura , Cimenos/química , Cimenos/farmacologia , Suplementos Nutricionais/análise , Alho/química , Imunidade Inata/genética , Imunidade nas Mucosas/efeitos dos fármacos , Muco/efeitos dos fármacos , Muco/microbiologia , Óleos Voláteis/classificação , Dourada/genética , Via Secretória/imunologia , Timol/química , Timol/farmacologia
8.
Gut ; 70(6): 1117-1129, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33177165

RESUMO

OBJECTIVE: The incidence of IBS increases following enteric infections, suggesting a causative role for microbial imbalance. However, analyses of faecal microbiota have not demonstrated consistent alterations. Here, we used metaproteomics to investigate potential associations between mucus-resident microbiota and IBS symptoms. DESIGN: Mucus samples were prospectively collected from sigmoid colon biopsies from patients with IBS and healthy volunteers, and their microbial protein composition analysed by mass spectrometry. Observations were verified by immunofluorescence, electron microscopy and real-time PCR, further confirmed in a second cohort, and correlated with comprehensive profiling of clinical characteristics and mucosal immune responses. RESULTS: Metaproteomic analysis of colon mucus samples identified peptides from potentially pathogenic Brachyspira species in a subset of patients with IBS. Using multiple diagnostic methods, mucosal Brachyspira colonisation was detected in a total of 19/62 (31%) patients with IBS from two prospective cohorts, versus 0/31 healthy volunteers (p<0.001). The prevalence of Brachyspira colonisation in IBS with diarrhoea (IBS-D) was 40% in both cohorts (p=0.02 and p=0.006 vs controls). Brachyspira attachment to the colonocyte apical membrane was observed in 20% of patients with IBS and associated with accelerated oro-anal transit, mild mucosal inflammation, mast cell activation and alterations of molecular pathways linked to bacterial uptake and ion-fluid homeostasis. Metronidazole treatment paradoxically promoted Brachyspira relocation into goblet cell secretory granules-possibly representing a novel bacterial strategy to evade antibiotics. CONCLUSION: Mucosal Brachyspira colonisation was significantly more common in IBS and associated with distinctive clinical, histological and molecular characteristics. Our observations suggest a role for Brachyspira in the pathogenesis of IBS, particularly IBS-D.


Assuntos
Proteínas de Bactérias/análise , Brachyspira/metabolismo , Infecções por Bactérias Gram-Negativas/epidemiologia , Síndrome do Intestino Irritável/microbiologia , Síndrome do Intestino Irritável/patologia , Muco/microbiologia , Adulto , Antibacterianos/farmacologia , Biópsia , Brachyspira/efeitos dos fármacos , Brachyspira/isolamento & purificação , Estudos de Casos e Controles , Colo Sigmoide/patologia , Diarreia/etiologia , Fezes/microbiologia , Feminino , Trânsito Gastrointestinal , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Negativas/patologia , Infecções por Bactérias Gram-Negativas/fisiopatologia , Humanos , Imunidade nas Mucosas , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Síndrome do Intestino Irritável/complicações , Síndrome do Intestino Irritável/fisiopatologia , Masculino , Mastócitos , Metronidazol/farmacologia , Pessoa de Meia-Idade , Muco/química , Prevalência , Estudos Prospectivos , Proteômica , Índice de Gravidade de Doença , Adulto Jovem
9.
BMC Microbiol ; 20(1): 301, 2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-33028252

RESUMO

BACKGROUND: The host-microbial commensalism can shape the innate immune responses in respiratory mucosa and nasal microbiome also modulates front-line immune mechanism in the nasal mucosa. Inhaled allergens encounter the host immune system first in the nasal mucosa, and microbial characteristics of nasal mucus directly impact the mechanisms of initial allergic responses in nasal epithelium. However, the roles of the nasal microbiome in allergic nasal mucosa remain uncertain. We sought to determine the distribution of nasal microbiomes in allergic nasal mucosa and elucidate the interplay between nasal microbiome Staphylococcus species and Th2 cytokines in allergic rhinitis (AR) models. RESULTS: Staphylococcus aureus (AR-SA) and S. epidermidis (AR-SE) were isolated from the nasal mucosa of patients with AR. The influence of nasal microbiome Staphylococcus species on allergic nasal mucosa was also tested with in vitro and in vivo AR models. Pyrosequencing data showed that colonization by S. epidermidis and S. aureus was more dominant in nasal mucus of AR subjects. The mRNA and protein levels of IL-33 and TSLP were significantly higher in AR nasal epithelial (ARNE) cells which were cultured from nasal mucosa of AR subjects, and exposure of ARNE cells to AR-SA reduced IL-33 mRNA and secreted protein levels. Particularly, ovalbumin-driven AR mice inoculated with AR-SA by intranasal delivery exhibited significantly reduced IL-33 in their nasal mucosa. In the context of these results, allergic symptoms and Th2 cytokine levels were significantly downregulated after intranasal inoculation of AR-SA in vivo AR mice. CONCLUSION: Colonization by Staphylococcus species was more dominant in allergic nasal mucosa, and nasal commensal S. aureus from subjects with AR mediates anti-allergic effects by modulating IL-33-dependent Th2 inflammation. The results demonstrate the role of host-bacterial commensalism in shaping human allergic inflammation.


Assuntos
Imunidade Inata , Mucosa Nasal/imunologia , Rinite Alérgica/imunologia , Staphylococcus aureus/imunologia , Staphylococcus epidermidis/imunologia , Simbiose/imunologia , Animais , Corynebacterium/crescimento & desenvolvimento , Citocinas/genética , Citocinas/imunologia , Modelos Animais de Doenças , Enterobacter aerogenes/crescimento & desenvolvimento , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Feminino , Expressão Gênica , Humanos , Interleucina-33/genética , Interleucina-33/imunologia , Camundongos Endogâmicos BALB C , Micrococcus luteus/crescimento & desenvolvimento , Muco/imunologia , Muco/microbiologia , Mucosa Nasal/microbiologia , Ovalbumina/administração & dosagem , Cultura Primária de Células , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Rinite Alérgica/induzido quimicamente , Rinite Alérgica/microbiologia , Rinite Alérgica/patologia , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus epidermidis/crescimento & desenvolvimento
10.
Respir Res ; 21(1): 222, 2020 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-32847538

RESUMO

Cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) are associated with acute and chronic bacterial infections of the lung. Excessive differentiation of basal cells to mucus-producing goblet cells can result in mucus hyperproduction and loss of mucociliary clearance in the airways of CF and COPD patients. Here, we aimed to investigate the effect of pathogen-associated molecular patterns (PAMPs) on the differentiation of human 3D bronchospheres. Primary human bronchial epithelial cells (HBECs) were differentiated to bronchospheres in the presence of bacterial flagellin and LPS and the synthetic Toll-like receptor (TLR) ligands Pam3CSK4 (TLR-2) and polyinosinic:polycytidylic acid (pIC, TLR-3). Electron and fluorescence microscopy showed that the differentiation of bronchospheres associated with the formation of lumina and appearance of cilia within 30 days after seeding. Incubation with flagellin resulted in a decreased formation of lumina and loss of cilia formation. Incubation with Pam3CSK, pIC, and LPS did not significantly affect formation of lumina and ciliation. Mucus production was strongly increased in response to flagellin and, to a lesser degree, in response to Pam3CSK4. Our results indicate that bacterial factors, such as flagellin, drive the differentiation of the respiratory epithelium towards mucus hyperproduction.


Assuntos
Brônquios/metabolismo , Flagelina/metabolismo , Depuração Mucociliar/fisiologia , Muco/metabolismo , Organoides/metabolismo , Mucosa Respiratória/metabolismo , Brônquios/microbiologia , Células Cultivadas , Flagelina/administração & dosagem , Humanos , Muco/microbiologia , Organoides/microbiologia , Organoides/ultraestrutura , Mucosa Respiratória/microbiologia , Mucosa Respiratória/ultraestrutura
11.
J Occup Environ Hyg ; 17(10): 437-446, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32776831

RESUMO

Pharmaceutical workers involved with the production of antimicrobial drugs are exposed to various antimicrobial chemicals in different steps of manufacturing such as grinding, sieving, compression, granulation, mixing, and filling. These exposures may lead to the development of multidrug resistance (MDR) in bacteria. Scientific reports on the occupational health hazard of pharmaceutical workers involved in manufacturing antibiotics are scarce. The present study aimed to compare the degree of bacterial resistance in pharmaceutical workers in India to that of individuals not involved in the pharmaceutical field. Twenty male workers from 5 local pharmaceutical companies and 20 male subjects not involved in the pharmaceutical field (non-pharmaceutical subjects) were randomly selected. Nasal fluid and mucus/cough specimens were collected from each subject and were cultured separately at 37 °C for 24 hr to obtain bacterial growth. The cultured species were then identified, isolated, and subjected to microbial sensitivity testing against 18 different antibiotics from 8 different groups by the disk diffusion method. Staphylococcus spp., Pseudomonas spp., and Escherichia coli were identified and isolated from the culture of nasal fluids and mucuses, respectively. All the isolated species of bacteria exhibited significant enhancement of the degree of MDR in pharmaceutical workers compared with non-pharmaceutical subjects. Workers with a longer working history had greater degree of antibiotic resistance and vice versa. It can be certainly considered that the exposure of pharmaceutical workers to antibiotic agents resulted in a high incidence of multidrug resistance. Effective steps should be taken to minimize inherent exposure of pharmaceutical workers to antibiotics during work to prevent antimicrobial drug resistance.


Assuntos
Antibacterianos/farmacologia , Indústria Farmacêutica , Farmacorresistência Bacteriana/efeitos dos fármacos , Exposição Ocupacional/efeitos adversos , Escherichia coli/efeitos dos fármacos , Humanos , Índia , Masculino , Testes de Sensibilidade Microbiana , Muco/microbiologia , Cavidade Nasal/microbiologia , Pseudomonas/efeitos dos fármacos , Medição de Risco , Staphylococcus/efeitos dos fármacos
12.
Nutrients ; 12(6)2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32545531

RESUMO

The Colonic Mucosal Barrier (CMB) is the site of interaction between the human body and the colonic microbiota. The mucus is the outer part of the CMB and is considered as the front-line defense of the colon. It separates the host epithelial lining from the colonic content, and it has previously been linked to health and diseases. In this study, we assessed the relationship between red meat and whole-grain intake and (1) the thickness of the colonic mucus (2) the expression of the predominant mucin gene in the human colon (MUC2). Patients referred to colonoscopy at the University Hospital of Southern Denmark- Sonderjylland were enrolled between June 2017 and December 2018, and lifestyle data was collected in a cross-sectional study design. Colonic biopsies, blood, urine, and fecal samples were collected. The colonic mucus and bacteria were visualized by immunostaining and fluorescence in situ hybridization techniques. We found a thinner mucus was associated with high red meat intake. Similarly, the results suggested a thinner mucus was associated with high whole-grain intake, albeit to a lesser extent than red meat. This is the first study assessing the association between red meat and whole-grain intake and the colonic mucus in humans. This study is approved by the Danish Ethics Committee (S-20160124) and the Danish Data Protecting Agency (2008-58-035). A study protocol was registered at clinical trials.gov under NCT04235348.


Assuntos
Colo/metabolismo , Dieta/métodos , Mucosa Intestinal/metabolismo , Carne Vermelha , Grãos Integrais , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Colo/microbiologia , Colo/patologia , Colonoscopia , Estudos Transversais , Dinamarca , Inquéritos sobre Dietas , Feminino , Humanos , Hibridização in Situ Fluorescente , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Microbiota , Pessoa de Meia-Idade , Mucina-2/genética , Mucina-2/metabolismo , Muco/metabolismo , Muco/microbiologia
13.
Nat Commun ; 11(1): 2097, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32350281

RESUMO

Astroviruses are a global cause of pediatric diarrhea, but they are largely understudied, and it is unclear how and where they replicate in the gut. Using an in vivo model, here we report that murine astrovirus preferentially infects actively secreting small intestinal goblet cells, specialized epithelial cells that maintain the mucus barrier. Consequently, virus infection alters mucus production, leading to an increase in mucus-associated bacteria and resistance to enteropathogenic E. coli colonization. These studies establish the main target cell type and region of the gut for productive murine astrovirus infection. They further define a mechanism by which an enteric virus can regulate the mucus barrier, induce functional changes to commensal microbial communities, and alter host susceptibility to pathogenic bacteria.


Assuntos
Infecções por Astroviridae/patologia , Infecções por Astroviridae/virologia , Astroviridae/fisiologia , Trato Gastrointestinal/patologia , Trato Gastrointestinal/virologia , Células Caliciformes/virologia , Muco/virologia , Animais , Células Epiteliais/patologia , Células Epiteliais/virologia , Escherichia coli/fisiologia , Feminino , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/ultraestrutura , Masculino , Camundongos Endogâmicos C57BL , Muco/microbiologia , Transcriptoma/genética , Replicação Viral/fisiologia , Eliminação de Partículas Virais/fisiologia
14.
Sci Rep ; 10(1): 4579, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32165709

RESUMO

The presence of bronchial mucus plugs (BMP) in children with Mycoplasma pneumoniae pneumonia (MPP) results in delayed clinical and radiographic resolution and long-standing pulmonary sequelae. The predictive factors associated with BMP formation remains poorly defined. Nomograms to predict BMP presence in children with MPP were proposed using a cohort of patients who underwent bronchoscopy intervention at Children's Hospital in Eastern China. Patients with MPP in an earlier period formed the training cohort (n = 872) for nomogram development, and those thereafter formed the validation cohort (n = 399) to confirmed model's performance. BMP in children with MPP were found in 196 (22.5%) and 91(22.8%) patients in the training and validation cohorts, respectively. The independent risk factors associated with BMP were age >5years (OR 2.06; 95% CI 1.43 to 2.98), higher IL-10 level (>10 ng/L, 2.19; 95% CI 1.46 to 3.28), higher IFN-γ level (>30 ng/L, 1.69; 95% CI 1.13 to 2.54), and presence of complication (3.43; 95% CI 1.45 to 8.09). Incorporating these 4 factors, the nomogram achieved good concordance indexes of 0.771(95% CI, 0.734-0.808) and 0.796 (95% CI, 0.744-0.848) in predicting BMP in the training and validation cohorts, respectively. The nomogram achieved an optimal prediction of BMP in children with MPP. Using this model, the risk of BMP formation would be determined, contributing to a rational therapeutic choice.


Assuntos
Muco/microbiologia , Mycoplasma pneumoniae/isolamento & purificação , Nomogramas , Pneumonia por Mycoplasma/diagnóstico , Broncoscopia , Criança , Pré-Escolar , Feminino , Humanos , Interferon gama/metabolismo , Interleucina-10/metabolismo , Masculino , Muco/imunologia , Pneumonia por Mycoplasma/imunologia , Fatores de Risco
15.
Gut Microbes ; 11(3): 364-380, 2020 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29494278

RESUMO

The pathogenesis of Crohn's disease (CD) is multifactorial and involves genetic susceptibility, environmental triggers and intestinal microbiota. Adherent-invasive Escherichia coli (AIEC) are flagellated bacteria more prevalent in CD patients than in healthy subjects and promote chronic intestinal inflammation. We aim at deciphering the role of flagella and flagellin modulation by intestinal conditions. AIEC flagellum expression is required for optimal adhesion to and invasion of intestinal epithelial cells. Interestingly, differential flagellin regulation was observed between commensal E. coli (HS) and AIEC (LF82) strains: flagellum expression by AIEC bacteria, in contrast to that of commensal E. coli, is enhanced under intestinal conditions (the presence of bile acids and mucins). Flagella are involved in the ability of the AIEC LF82 strain to cross a mucus layer in vitro and in vivo, conferring a selective advantage in penetrating the mucus layer and reaching the epithelial surface. In a CEABAC10 mouse model, a non-motile mutant (LF82-ΔfliC) exhibits reduced colonization that is restored by a dextran sodium sulfate treatment that alters mucus layer integrity. Moreover, a mutant that continuously secretes flagellin (LF82-ΔflgM) triggers a stronger inflammatory response than the wild-type strain, and the mutant's ability to colonize the CEABAC10 mouse model is decreased. Overexpression of flagellin in bacteria in contact with epithelial cells can be detrimental to their virulence by inducing acute inflammation that enhances AIEC clearance. AIEC pathobionts must finely modulate flagellum expression during the infection process, taking advantage of their specific virulence gene regulation to improve their adaptability and flexibility within the gut environment.


Assuntos
Aderência Bacteriana , Escherichia coli/fisiologia , Flagelos/metabolismo , Intestinos/microbiologia , Animais , Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células CACO-2 , Contagem de Colônia Microbiana , Doença de Crohn/microbiologia , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/patogenicidade , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Flagelos/genética , Flagelina/genética , Flagelina/metabolismo , Regulação Bacteriana da Expressão Gênica , Células HT29 , Humanos , Mucosa Intestinal/microbiologia , Intestinos/química , Camundongos , Camundongos Endogâmicos C57BL , Muco/microbiologia , Mutação , Fenótipo
16.
Am J Respir Crit Care Med ; 201(6): 661-670, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31765597

RESUMO

Rationale: Non-cystic fibrosis bronchiectasis is characterized by airway mucus accumulation and sputum production, but the role of mucus concentration in the pathogenesis of these abnormalities has not been characterized.Objectives: This study was designed to: 1) measure mucus concentration and biophysical properties of bronchiectasis mucus; 2) identify the secreted mucins contained in bronchiectasis mucus; 3) relate mucus properties to airway epithelial mucin RNA/protein expression; and 4) explore relationships between mucus hyperconcentration and disease severity.Methods: Sputum samples were collected from subjects with bronchiectasis, with and without chronic erythromycin administration, and healthy control subjects. Sputum percent solid concentrations, total and individual mucin concentrations, osmotic pressures, rheological properties, and inflammatory mediators were measured. Intracellular mucins were measured in endobronchial biopsies by immunohistochemistry and gene expression. MUC5B (mucin 5B) polymorphisms were identified by quantitative PCR. In a replication bronchiectasis cohort, spontaneously expectorated and hypertonic saline-induced sputa were collected, and mucus/mucin concentrations were measured.Measurements and Main Results: Bronchiectasis sputum exhibited increased percent solids, total and individual (MUC5B and MUC5AC) mucin concentrations, osmotic pressure, and elastic and viscous moduli compared with healthy sputum. Within subjects with bronchiectasis, sputum percent solids correlated inversely with FEV1 and positively with bronchiectasis extent, as measured by high-resolution computed tomography, and inflammatory mediators. No difference was detected in MUC5B rs35705950 SNP allele frequency between bronchiectasis and healthy individuals. Hypertonic saline inhalation acutely reduced non-cystic fibrosis bronchiectasis mucus concentration by 5%.Conclusions: Hyperconcentrated airway mucus is characteristic of subjects with bronchiectasis, likely contributes to disease pathophysiology, and may be a target for pharmacotherapy.


Assuntos
Bronquiectasia/tratamento farmacológico , Bronquiectasia/fisiopatologia , Eritromicina/uso terapêutico , Muco/química , Sistema Respiratório/fisiopatologia , Escarro/química , Idoso , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Muco/microbiologia , Queensland , Escarro/microbiologia
17.
Clin Respir J ; 14(2): 140-147, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31758867

RESUMO

INTRODUCTION: The characteristics of Allergic Bronchopulmonary Aspergillosis (ABPA) based on its radiological classification is still unclear. OBJECTIVES: To investigate the clinical significances of ABPA patients with central bronchiectasis (ABPA-CB) by different radiological classifications of mucus plugs. METHODS: ABPA-CB patients from a pulmonary hospital between 2008 and 2015 were retrospectively included and analysed. According to the chest imaging in their first visit to physician, the ABPA-CB patients were divided into two groups based on the presence of high-attenuation mucus (HAM) or low-attenuation mucus (LAM). The primary endpoint was ABPA relapse within 1 year since the glucocorticoid withdrawal. The relationship between the imaging findings and the clinical prognosis was illuminated. RESULTS: A total of 125 ABPA patients were analysed in this study. Compared to the LAM group, the HAM group presented higher blood eosinophil cells counts, higher rates of Aspergillus detection isolated in sputum and expectoration of brownish-black mucus plugs, more affected lobes and segments, poorer pulmonary function and higher rate of relapse. CONCLUSIONS: The clinical characteristics and prognosis of ABPA-CB patients are closely related to its radiological phenotype of mucus plugs in the central bronchiectasis. Clinicians should promote a diversity of personalized treatments for different patients with different radiological characteristics.


Assuntos
Aspergillus/isolamento & purificação , Bronquiectasia/etiologia , Broncoscopia/métodos , Muco/microbiologia , Aspergilose Pulmonar/complicações , Tomografia Computadorizada por Raios X/métodos , Adulto , Bronquiectasia/classificação , Bronquiectasia/diagnóstico , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Aspergilose Pulmonar/diagnóstico , Aspergilose Pulmonar/microbiologia , Estudos Retrospectivos
18.
Sci Rep ; 9(1): 15683, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31666581

RESUMO

Akkermansia muciniphila utilises colonic mucin as its substrate. Abundance is reduced in ulcerative colitis (UC), as is the relative proportion of sulphated mucin in the mucus gel layer (MGL). It is unknown if these phenomena are related, however reduced sulphated mucins could contribute to reduced abundance, owing to a lack of substrate. The aim of this study was to quantify A. muciniphila within the MGL and to relate these findings with markers of inflammation and the relative proportion of sulphomucin present. Colonic biopsies and mucus brushings were obtained from 20 patients with active UC (AC), 14 with quiescent UC (QUC) and 20 healthy controls (HC). A. muciniphila abundance was determined by RT-PCR. High iron diamine alcian-blue staining was performed for histological analysis. Patients with AC had reduced abundance of A. muciniphila compared to HC and QUC. A positive association was found between A. muciniphila abundance and higher percentage of sulphated mucin (ρ 0.546, p = 0.000). Lower abundances of A. muciniphila correlated with higher inflammatory scores (ρ = 0.294 (p = 0.001)). This study confirms an inverse relationship between A. muciniphila and inflammation and a positive association between A. muciniphila abundance and percentage of sulfated mucin in the MGL.


Assuntos
Colite Ulcerativa/microbiologia , Inflamação/genética , Mucinas/metabolismo , Verrucomicrobia/metabolismo , Adolescente , Adulto , Akkermansia , Biópsia , Colite Ulcerativa/metabolismo , Colite Ulcerativa/patologia , Colo/metabolismo , Colo/microbiologia , Feminino , Voluntários Saudáveis , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Masculino , Pessoa de Meia-Idade , Mucinas/isolamento & purificação , Muco/metabolismo , Muco/microbiologia , Verrucomicrobia/patogenicidade , Adulto Jovem
19.
Nat Microbiol ; 4(12): 2146-2154, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31611643

RESUMO

A slimy, hydrated mucus gel lines all wet epithelia in the human body, including the eyes, lungs, and gastrointestinal and urogenital tracts. Mucus forms the first line of defence while housing trillions of microorganisms that constitute the microbiota1. Rarely do these microorganisms cause infections in healthy mucus1, suggesting that mechanisms exist in the mucus layer that regulate virulence. Using the bacterium Pseudomonas aeruginosa and a three-dimensional (3D) laboratory model of native mucus, we determined that exposure to mucus triggers downregulation of virulence genes that are involved in quorum sensing, siderophore biosynthesis and toxin secretion, and rapidly disintegrates biofilms-a hallmark of mucosal infections. This phenotypic switch is triggered by mucins, which are polymers that are densely grafted with O-linked glycans that form the 3D scaffold inside mucus. Here, we show that isolated mucins act at various scales, suppressing distinct virulence pathways, promoting a planktonic lifestyle, reducing cytotoxicity to human epithelia in vitro and attenuating infection in a porcine burn model. Other viscous polymer solutions lack the same effect, indicating that the regulatory function of mucin does not result from its polymeric structure alone. We identify that interactions with P. aeruginosa are mediated by mucin-associated glycans (mucin glycans). By isolating glycans from the mucin backbone, we assessed the collective activity of hundreds of complex structures in solution. Similar to their grafted counterparts, free mucin glycans potently regulate bacterial phenotypes even at relatively low concentrations. This regulatory function is likely dependent on glycan complexity, as monosaccharides do not attenuate virulence. Thus, mucin glycans are potent host signals that 'tame' microorganisms, rendering them less harmful to the host.


Assuntos
Interações Hospedeiro-Patógeno , Mucinas/química , Muco/microbiologia , Polissacarídeos/química , Pseudomonas aeruginosa/patogenicidade , Animais , Biofilmes , Queimaduras/microbiologia , Células Epiteliais/microbiologia , Feminino , Células HT29 , Humanos , Muco/química , Pseudomonas aeruginosa/efeitos dos fármacos , Percepção de Quorum , Suínos , Virulência/genética , Ferimentos e Lesões/microbiologia
20.
Expert Rev Gastroenterol Hepatol ; 13(10): 963-976, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31603356

RESUMO

Introduction: Inflammatory bowel diseases (IBD) are on the rise worldwide. This review covers the current concepts of the etiology of Crohn´s disease and ulcerative colitis by focusing on an unbalanced interaction between the intestinal microbiota and the mucosal barrier. Understanding these issues is of paramount importance for the development of targeted therapies aiming at the disease cause.Area covered: Gut microbiota alterations and a dysfunctional intestinal mucosa are associated with IBD. Here we focus on specific defense structures of the mucosal barrier, namely antimicrobial peptides and the mucus layer, which keep the gut microbiota at a distance under healthy conditions and are defective in IBD.Expert commentary: The microbiology of both forms of IBD is different but characterized by a reduced bacterial diversity and richness. Abundance of certain bacterial species is altered, and the compositional changes are related to disease activity. In IBD the mucus layer above the epithelium is contaminated by bacteria and the immune reaction is dominated by the antibacterial response. Human genetics suggest that many of the basic deficiencies in the mucosal response, due to Paneth cell, defensin and mucus defects, are primary. Nutrition may also be important but so far there is no therapy targeting the mucosal barrier.


Assuntos
Colite Ulcerativa/microbiologia , Doença de Crohn/microbiologia , Microbioma Gastrointestinal , Mucosa Intestinal/microbiologia , Animais , Antibacterianos/uso terapêutico , Peptídeos Catiônicos Antimicrobianos/imunologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/imunologia , Colite Ulcerativa/metabolismo , Doença de Crohn/tratamento farmacológico , Doença de Crohn/imunologia , Doença de Crohn/metabolismo , Disbiose , Microbioma Gastrointestinal/efeitos dos fármacos , Interações Hospedeiro-Patógeno , Humanos , Imunidade nas Mucosas , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Muco/imunologia , Muco/metabolismo , Muco/microbiologia , Probióticos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA