Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 265(Pt 2): 130933, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38508554

ABSTRACT

Glucans, a polysaccharide naturally present in the yeast cell wall that can be obtained from side streams generated during the fermentation process, have gained increasing attention for their potential as a skin ingredient. Therefore, this study focused on the extraction method to isolate and purify water-insoluble glucans from two different Saccharomyces cerevisiae strains: an engineered strain obtained from spent yeast in an industrial fermentation process and a wild strain produced through lab-scale fermentation. Two water-insoluble extracts with a high glucose content (> 90 %) were achieved and further subjected to a chemical modification using carboxymethylation to improve their water solubility. All the glucans' extracts, water-insoluble and carboxymethylated, were structurally and chemically characterized, showing almost no differences between both yeast-type strains. To ensure their safety for skin application, a broad safety assessment was undertaken, and no cytotoxic effect, immunomodulatory capacity (IL-6 and IL-8 regulation), genotoxicity, skin sensitization, and impact on the skin microbiota were observed. These findings highlight the potential of glucans derived from spent yeast as a sustainable and safe ingredient for cosmetic and skincare formulations, contributing to the sustainability and circular economy.


Subject(s)
Glucans , Saccharomyces cerevisiae , Glucans/chemistry , Saccharomyces cerevisiae/chemistry , Polysaccharides/chemistry , Water
2.
Int J Mol Sci ; 24(3)2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36768709

ABSTRACT

Cannabidiol (CBD) and cannabigerol (CBG) are two pharmacologically active phytocannabinoids of Cannabis sativa L. Their antimicrobial activity needs further elucidation, particularly for CBG, as reports on this cannabinoid are scarce. We investigated CBD and CBG's antimicrobial potential, including their ability to inhibit the formation and cause the removal of biofilms. Our results demonstrate that both molecules present activity against planktonic bacteria and biofilms, with both cannabinoids removing mature biofilms at concentrations below the determined minimum inhibitory concentrations. We report for the first time minimum inhibitory and lethal concentrations for Pseudomonas aeruginosa and Escherichia coli (ranging from 400 to 3180 µM), as well as the ability of cannabinoids to inhibit Staphylococci adhesion to keratinocytes, with CBG demonstrating higher activity than CBD. The value of these molecules as preservative ingredients for cosmetics was also assayed, with CBG meeting the USP 51 challenge test criteria for antimicrobial effectiveness. Further, the exact formulation showed no negative impact on skin microbiota. Our results suggest that phytocannabinoids can be promising topical antimicrobial agents when searching for novel therapeutic candidates for different skin conditions. Additional research is needed to clarify phytocannabinoids' mechanisms of action, aiming to develop practical applications in dermatological use.


Subject(s)
Cannabidiol , Cannabinoids , Cannabis , Cannabidiol/pharmacology , Cannabinoids/pharmacology , Skin
3.
Microb Ecol ; 86(1): 86-96, 2023 Jul.
Article in English | MEDLINE | ID: mdl-35809121

ABSTRACT

Skin harbors an important microbial ecosystem - the skin microbiota that is in homeostasis with its host and is beneficial for human health. Cosmetic products have the potential to interfere with this microbial community; therefore their impact should be assessed. The aim of this review is to highlight the importance of skin microbiota in the cosmetic industry. Several studies determined that cosmetic ingredients have the potential to disrupt the skin microbiota equilibrium leading to the development of skin diseases and dysregulation of immune response. These studies led their investigation by using different methodologies and models, concluding that methods must be chosen according to the aim of the study, the skin site to be evaluated, and the target population of the cosmetics. Overall, it is crucial to test the impact of cosmetics in the skin microbiota and to stablish standard procedures, as well as specific criteria that allow to classify a cosmetic product as skin microbiota friendly.


Subject(s)
Cosmetics , Host Microbial Interactions , Microbiota , Skin , Humans , Cosmetics/pharmacology , Homeostasis , Microbiota/drug effects , Skin/microbiology , Host Microbial Interactions/drug effects , Industry/standards , Industry/trends
4.
J Infect Dis ; 226(12): 2226-2237, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36173814

ABSTRACT

BACKGROUND: Helicobacter pylori infection induces cellular phenotypes relevant for cancer progression, namely cell motility and invasion. We hypothesized that the extracellular matrix (ECM) could be involved in these deleterious effects. METHODS: Microarrays were used to uncover ECM interactors in cells infected with H. pylori. LAMC2, encoding laminin γ2, was selected as a candidate gene and its expression was assessed in vitro and in vivo. The role of LAMC2 was investigated by small interference RNA (siRNA) combined with a set of functional assays. Laminin γ2 and E-cadherin expression patterns were evaluated in gastric cancer cases. RESULTS: Laminin γ2 was found significantly overexpressed in gastric cancer cells infected with H. pylori. This finding was validated in vitro by infection with clinical isolates and in vivo by using gastric biopsies of infected and noninfected individuals. We showed that laminin γ2 overexpression is dependent on the bacterial type IV secretion system and on the CagA. Functionally, laminin γ2 promotes cell invasion and resistance to apoptosis, through modulation of Src, JNK, and AKT activity. These effects were abrogated in cells with functional E-cadherin. CONCLUSIONS: These data highlight laminin γ2 and its downstream effectors as potential therapeutic targets, and the value of H. pylori eradication to delay gastric cancer onset and progression.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Helicobacter pylori/genetics , Laminin/metabolism , Helicobacter Infections/microbiology , Cell Line, Tumor , Cadherins/metabolism , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
5.
Int J Mol Sci ; 21(3)2020 Feb 07.
Article in English | MEDLINE | ID: mdl-32046034

ABSTRACT

Large numbers of well-characterized clinical samples are fundamental to establish relevant associations between the microbiota and disease. Formalin-fixed and paraffin-embedded (FFPE) tissues are routinely used and are widely available clinical materials. Since current approaches to study the microbiota are based on next-generation sequencing (NGS) targeting the bacterial 16S rRNA gene, our aim was to evaluate the feasibility of FFPE gastric tissues for NGS-based microbiota characterization. Analysis of sequencing data revealed the presence of bacteria in the paraffin control. After the subtraction of the operational taxonomic units (OTUs) present in the paraffin control to the FFPE tissue sample dataset, we evaluated the microbiota profiles between paired FFPE and frozen gastric tissues, and between different times of archiving. Compared with frozen gastric tissues, we detected a lower number of OTUs in the microbiota of paired FFPE tissues, regardless of the time of archiving. No major differences in microbial diversity were identified, but taxonomic variation in the relative abundance of phyla and orders was observed between the two preservation methods. This variation was also evident in each case and throughout the times of FFPE archiving. The use of FFPE tissues for NGS-based microbiota characterization should be considered carefully, as biases can be introduced by the embedding process and the time of tissue archiving.


Subject(s)
DNA Barcoding, Taxonomic/methods , High-Throughput Nucleotide Sequencing/methods , Microbiota , Paraffin Embedding/methods , Stomach/microbiology , Tissue Fixation/methods , Fixatives/adverse effects , Formaldehyde/adverse effects , Genome, Bacterial , Humans , RNA, Ribosomal, 16S/genetics , Stomach/cytology
6.
Front Microbiol ; 10: 1277, 2019.
Article in English | MEDLINE | ID: mdl-31244801

ABSTRACT

The amount of host DNA poses a major challenge to metagenome analysis. However, there is no guidance on the levels of host DNA, nor on the depth of sequencing needed to acquire meaningful information from whole metagenome sequencing (WMS). Here, we evaluated the impact of a wide range of amounts of host DNA and sequencing depths on microbiome taxonomic profiling using WMS. Synthetic samples with increasing levels of host DNA were created by spiking DNA of a mock bacterial community, with DNA from a mouse-derived cell line. Taxonomic analysis revealed that increasing proportions of host DNA led to decreased sensitivity in detecting very low and low abundant species. Reduction of sequencing depth had major impact on the sensitivity of WMS for profiling samples with 90% host DNA, increasing the number of undetected species. Finally, analysis of simulated datasets with fixed depth of 10 million reads confirmed that microbiome profiling becomes more inaccurate as the level of host DNA increases in a sample. In conclusion, samples with high amounts of host DNA coupled with reduced sequencing depths, decrease WMS coverage for characterization of the microbiome. This study highlights the importance of carefully considering these aspects in the design of WMS experiments to maximize microbiome analyses.

7.
Adv Exp Med Biol ; 1149: 195-210, 2019.
Article in English | MEDLINE | ID: mdl-31016631

ABSTRACT

After a long period during which the stomach was considered as an organ where microorganisms could not thrive, Helicobacter pylori was isolated in vitro from gastric biopsies, revolutionising the fields of Microbiology and Gastroenterology. Since then, and with the introduction of high-throughput sequencing technologies that allowed deep characterization of microbial communities, a growing body of knowledge has shown that the stomach contains a diverse microbial community, which is different from that of the oral cavity and of the intestine. Gastric cancer is a heterogeneous disease that is the end result of a cascade of events arising in a small fraction of patients colonized with H. pylori. In addition to H. pylori infection and to multiple host and environmental factors that influence disease development, alterations to the composition and function of the normal gastric microbiome, also known as dysbiosis, may also contribute to malignancy. Chronic inflammation of the mucosa in response to H. pylori may alter the gastric environment, paving the way to the growth of a dysbiotic gastric bacterial community. This dysbiotic microbiome may promote the development of gastric cancer by sustaining inflammation and/or inducing genotoxicity. This chapter summarizes what is known about the gastric microbiome in the context of H. pylori-associated gastric cancer, introducing the emerging dimension of the microbiome into the pathogenesis of this highly incident and deadly disease.


Subject(s)
Gastrointestinal Microbiome , Helicobacter Infections , Helicobacter pylori , Dysbiosis , Gastric Mucosa/microbiology , Gastrointestinal Microbiome/physiology , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Humans
8.
BMC Mol Biol ; 19(1): 6, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29879907

ABSTRACT

BACKGROUND: Gastric cancer is the third leading cause of cancer-related mortality worldwide. Recently, it has been demonstrated that gastric cancer cells display a specific miRNA expression profile, with increasing evidence of the role of miRNA-9 in this disease. miRNA-9 upregulation has been shown to influence the expression of E-cadherin-encoding gene, triggering cell motility and invasiveness. RESULTS: In this study, we designed LNA anti-miRNA oligonucleotides with a complementary sequence to miRNA-9 and tested their properties to both detect and silence the target miRNA. We could identify and visualize the in vitro uptake of low-dosing LNA-based anti-miRNA oligonucleotides without any carrier or transfection agent, as early as 2 h after the addition of the oligonucleotide sequence to the culture medium. Furthermore, we were able to assess the silencing potential of miRNA-9, using different LNA anti-miRNA oligonucleotide designs, and to observe its subsequent effect on E-cadherin expression. CONCLUSIONS: The administration of anti-miRNA sequences even at low-doses, rapidly repressed the target miRNA, and influenced the expression of E-cadherin by significantly increasing its levels.


Subject(s)
Cadherins/genetics , MicroRNAs/antagonists & inhibitors , Oligonucleotides/pharmacology , Stomach Neoplasms/genetics , Antigens, CD , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism
9.
Gastrointest Endosc ; 87(6): 1566-1575, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29454568

ABSTRACT

BACKGROUND AND AIMS: The time course for the development of clinically significant hereditary diffuse gastric cancer (HDGC) is unpredictable. Little is known about the progression from preclinical, indolent lesions to widely invasive, aggressive phenotypes. Gastroendoscopy often fails to detect early lesions, and risk-reducing/prophylactic total gastrectomy (PTG) is the only curative approach. We present an HDGC family with early-onset disease in which clinical and histologic findings provided insight into the understanding of different HDGC phenotypes. METHODS: The proband was diagnosed at age 18 years with widely invasive, metastatic DGC. CDH1 genetic testing identified a pathogenic, germline CDH1 variant (c.1901C>T, p.Ala634Val). Thirty family members were tested, and 15 CDH1 carriers were identified. RESULTS: Six family members had PTG, with negative preoperative workup. The proband's 14-year-old sister is the youngest patient, reported to date, to have PTG after negative preoperative biopsy sampling. Intramucosal HDGC foci were detected in all PTG specimens (1-33). In contrast to the "indolent" phenotype of these foci, the aggressive DGC from the proband showed pleomorphic cells, absent E-cadherin expression, increased proliferation (Ki-67 index), and activation of oncogenic events (p53, pSrc and pStat3 overexpression). All family members had Helicobacter pylori gastritis. Cag-A-positive strains were detected in all specimens, except in the proband's sister. CONCLUSIONS: HDGC is a heterogeneous disease regarding clinical behavior, endoscopic findings, histopathologic features, and immunophenotypic/molecular profile. The presence of bizarre, pleomorphic cells in endoscopic biopsy specimens is suggestive of advanced disease and should prompt clinical intervention. The involvement of a full multidisciplinary team is essential for the management of these patients.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Lobular/pathology , Neoplastic Syndromes, Hereditary/pathology , Stomach Neoplasms/pathology , Adolescent , Adult , Age of Onset , Aged, 80 and over , Antigens, CD , Breast Neoplasms/genetics , Cadherins/genetics , Carcinoma, Lobular/genetics , Family , Female , Gastrectomy , Gastritis/complications , Gastritis/microbiology , Gastroscopy , Genetic Predisposition to Disease , Germ-Line Mutation , Helicobacter Infections/complications , Helicobacter pylori , Humans , Ki-67 Antigen/metabolism , Male , Middle Aged , Mutation, Missense , Neoplastic Syndromes, Hereditary/complications , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/prevention & control , Pedigree , Phenotype , Prophylactic Surgical Procedures , Stomach Neoplasms/complications , Stomach Neoplasms/genetics , Stomach Neoplasms/prevention & control , Tumor Suppressor Protein p53/metabolism , Young Adult
10.
Gut ; 67(2): 226-236, 2018 02.
Article in English | MEDLINE | ID: mdl-29102920

ABSTRACT

OBJECTIVE: Gastric carcinoma development is triggered by Helicobacter pylori. Chronic H. pylori infection leads to reduced acid secretion, which may allow the growth of a different gastric bacterial community. This change in the microbiome may increase aggression to the gastric mucosa and contribute to malignancy. Our aim was to evaluate the composition of the gastric microbiota in chronic gastritis and in gastric carcinoma. DESIGN: The gastric microbiota was retrospectively investigated in 54 patients with gastric carcinoma and 81 patients with chronic gastritis by 16S rRNA gene profiling, using next-generation sequencing. Differences in microbial composition of the two patient groups were assessed using linear discriminant analysis effect size. Associations between the most relevant taxa and clinical diagnosis were validated by real-time quantitative PCR. Predictive functional profiling of microbial communities was obtained with PICRUSt. RESULTS: The gastric carcinoma microbiota was characterised by reduced microbial diversity, by decreased abundance of Helicobacter and by the enrichment of other bacterial genera, mostly represented by intestinal commensals. The combination of these taxa into a microbial dysbiosis index revealed that dysbiosis has excellent capacity to discriminate between gastritis and gastric carcinoma. Analysis of the functional features of the microbiota was compatible with the presence of a nitrosating microbial community in carcinoma. The major observations were confirmed in validation cohorts from different geographic origins. CONCLUSIONS: Detailed analysis of the gastric microbiota revealed for the first time that patients with gastric carcinoma exhibit a dysbiotic microbial community with genotoxic potential, which is distinct from that of patients with chronic gastritis.


Subject(s)
Bacteria , Carcinoma/microbiology , Dysbiosis/microbiology , Gastritis/microbiology , Gastrointestinal Microbiome , Helicobacter Infections/microbiology , Stomach Neoplasms/microbiology , Stomach/microbiology , Adult , Aged , Bacteria/genetics , Bacteria/metabolism , Chronic Disease , Female , Gastric Mucosa/microbiology , Helicobacter pylori , Humans , Male , Middle Aged , Nitrosation , RNA, Ribosomal, 16S/analysis , Retrospective Studies
11.
Toxins (Basel) ; 8(5)2016 05 05.
Article in English | MEDLINE | ID: mdl-27164143

ABSTRACT

Helicobacter pylori is the major triggering factor for gastric carcinoma, but only a small proportion of infected patients develop this disease. Differences in virulence observed among H. pylori strains, namely in the vacuolating cytotoxin vacA gene, may contribute to this discrepancy. Infection with vacA s1, i1 and m1 strains increases the risk for progression of gastric premalignant lesions and for gastric carcinoma. However, in East Asian countries most of the H. pylori strains are vacA s1, regardless of the patients' clinical status, and the significance of the vacA i1 and m1 genotypes for gastric carcinoma in this geographic area remains to be fully elucidated. The aim of the present study was to investigate this relationship in 290 patients from Macau, China. Using very sensitive and accurate genotyping methods, we detected infection with vacA i1 and with vacA m1 strains in, respectively, 85.2% and 52.6% of the patients that were infected with single genotypes. The prevalence of cagA-positive strains was 87.5%. No significant associations were observed between vacA genotypes or cagA and gastric carcinoma. It is worth noting that 37.5% of the infected patients had coexistence of H. pylori strains with different vacA genotypes. Additional studies directed to other H. pylori virulence factors should be performed to identify high risk patients in East Asia.


Subject(s)
Bacterial Proteins/genetics , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Stomach Neoplasms/microbiology , Aged , Bacterial Toxins/genetics , China/epidemiology , Chronic Disease , DNA, Bacterial/genetics , Female , Gastritis/epidemiology , Gastritis/pathology , Genes, Bacterial , Genotype , Helicobacter Infections/epidemiology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Humans , Male , Middle Aged , Stomach Neoplasms/epidemiology , Stomach Neoplasms/pathology , Virulence
12.
J Infect Dis ; 213(11): 1767-76, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26802142

ABSTRACT

Helicobacter pylori colonizes the human stomach and increases the risk for peptic ulcer disease and gastric carcinoma. H. pylori upregulates the expression and activity of several matrix metalloproteinases (MMPs) in cell lines and in the gastric mucosa. The aim of this study was to explore the mechanisms leading to upregulation of MMP10 in gastric epithelial cells induced by H. pylori Infection of gastric cells with H. pylori led to an increase in levels of MMP-10 messenger RNA, protein secretion, and activity. cagA knockout mutants or CagA phosphorylation-defective mutants failed to increase MMP10 expression. These results were confirmed in infection experiments with clinical isolates with known cagA status and in human gastric biopsy specimens. Treatment of cells with chemical inhibitors of the receptor tyrosine kinase EGFR and the kinase Src abrogated H. pylori-induced MMP10 expression. Inhibitors of ERK1/2 and JNK kinases abolished and significantly decreased H. pylori-induced MMP10 expression, respectively, whereas inhibition of the kinase p38 had no effect. Finally, inhibition of MMP10 expression by small interfering RNA led to a decrease in the gastric cell-invasive phenotype mediated by the infection. In conclusion, CagA-positive H. pylori strains stimulate MMP10 expression. MMP-10 modulation occurs via EGFR activation in a process that involves Src, ERK, and JNK pathways. MMP-10 may be implicated in H. pylori-mediated extracellular matrix remodeling.


Subject(s)
ErbB Receptors/metabolism , Gastric Mucosa/enzymology , Helicobacter pylori/pathogenicity , MAP Kinase Signaling System , Matrix Metalloproteinase 10/metabolism , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Line , Cell Line, Tumor , Enzyme Activation , Female , Gastric Mucosa/microbiology , Humans , MAP Kinase Kinase 4/metabolism , Male , Middle Aged , Virulence Factors/metabolism
13.
J Infect Dis ; 213(4): 669-73, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26401027

ABSTRACT

Heterogeneity at the Helicobacter pylori cagA gene promoter region has been linked to variation in CagA expression and gastric histopathology. Here, we characterized the cagA promoter and expression in 46 H. pylori strains from Portugal. Our results confirm the relationship between cagA promoter region variation and protein expression originally observed in strains from Colombia. We observed that individuals with intestinal metaplasia were all infected with H. pylori strains containing a specific cagA motif. Additionally, we provided novel functional evidence that strain-specific sequences in the cagA promoter region and CagA expression levels influence interleukin 8 secretion by the host gastric epithelial cells.


Subject(s)
Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Gene Expression , Helicobacter pylori/genetics , Helicobacter pylori/immunology , Interleukin-8/metabolism , Promoter Regions, Genetic , Adult , Aged , Case-Control Studies , Epithelial Cells/immunology , Epithelial Cells/microbiology , Female , Genetic Variation , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/isolation & purification , Humans , Male , Middle Aged , Portugal , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...