INTRODUCTION
For several centuries, the production of wine, bread, cheese, and multiple types of fermented food from Europe to Asia relies on the use of fungal strains (1). Over time, academic laboratories and many private companies have fostered research on the diversity of these different yeast strains and their potential in food production through fermentation and aroma production. The repertoire of these strains is now very large and offers a considerable quantity of physiological properties that can be applied in various food processes. Among fungi, the most well-known fungus is the yeast
MATERIALS AND METHODS
Cell culture
Human enterocyte-like Caco-2 (ATCC, Virginia, USA) cells were grown in Dulbecco’s modified Eagle’s medium (DMEM) (GIBCO Fischer-Scientific, France) supplemented with 4.5 g/L D-glucose (Sigma-Aldrich, France), pyruvate (Sigma-Aldrich, France), 10% heat-inactivated fetal calf serum (FBS, Eurobio Scientific), 1% nonessential amino acids (GIBCO Fischer-Scientific, France), 1% L-glutamine (Sigma-Aldrich, France), 50 IU/mL penicillin (Sigma-Aldrich, France), and 50 µg/mL streptomycin (Gibco, Fischer-Scientific France) at 37°C in a 5% CO2 atmosphere. The growth medium was changed every day with fresh medium and without the addition of antibiotics for the last 24 h of incubation prior to performing the adhesion assays.
HT29-MTX (Micalis Institut) from the human colon was grown in DMEM supplemented with 4.5 g/L D-glucose (Sigma-Aldrich, France), pyruvate (Sigma-Aldrich, France), 10% heat-inactivated (Eurobio Scientific), 50 IU/mL penicillin (Sigma-Aldrich, France), and 50 ug/mL streptomycin (Gibco Fischer-Scientific France) at 37°C in a 5% CO2 atmosphere. The growth medium was changed every day with fresh medium and without the addition of antibiotics for the last 24 h of incubation prior to performing the adhesion assays.
The HT-29 cell line (passages P3 to P10) was obtained from the supplier European Collection of Authenticated Cell Cultures. HT29 cells were cultured in DMEM containing 200 mM glutamine (Gibco, Fischer-Scientific, France), 50 IU/mL penicillin (Sigma-Aldrich, France), 50 µg/mL streptomycin (Gibco, Fischer-Scientific, France), and 10% heat inactivated FBS (Eurobio Scientific) at 37°C in a 5% CO2 atmosphere. The growth medium was changed every 2 days with fresh medium. At a confluence of approximately 80%, HT29 cells were treated with trypsin-EDTA (Gibco Fischer-Scientific, France) and distributed in 24-well plates (Dutscher, France) with 50,000 cells/well for the following 7 days.
Preparation of fungi
All strains were supplied by the company DuPont Nutrition and Biosciences.
Fungal adhesion on Caco2 and HT29-MTX cells
Caco-2 and HT29-MTX cells were seeded at 5.104 cells per well in 24-well plates (for yeasts) and incubated at 37°C in a 5% CO2 atmosphere. After 2 days of culture, the medium was changed every other day until coincubation. Cells were ready to use 15–21 days after confluence for Caco-2 cells and 15–18 days after confluence for HT29-MTX cells when they started to produce mucus. For coincubation, 104 CFU of the yeast strains or 2.106 spores were used per well. Independent experiments were repeated three to six times depending on the sample tested.
Quantitative assessment of adhesion
Yeast strain adherence to cultured enterocytes was assayed by the CFU method. After 1 h of coincubation, enterocytes were washed five times with 0.5 mL of PBS (Gibco, Fischer-Scientific, France), lysed with 0.5 mL of PBS (Gibco, Fischer-Scientific, France) with 0.1% Triton X-100 (Sigma-Aldrich, France), scraped if necessary, and collected for CFU determination after serial dilution on YEPD agar plates (yeast culture medium) (Gibco, Fischer-Scientific, France). The adhesion percentage was determined by calculating the ratio between the CFU determined in the inoculum and in the lysate.
DNA extraction
Samples were disrupted by using Precellys (Bertin Instruments, USA) with 2 cycles of 20 s at 10,000 rpm. Fifty microliters of 10% SDS were added to the tube, and the tubes were mixed by vortexing and incubated for 30 min at 65°C. After incubation, the samples were disrupted a second time with 1 cycle of 20 s at 10,000 rpm. Then, 200 µL of 5 M K-acetate (Sigma-Aldrich, France) was added to the sample, gently mixed, and incubated overnight at 4°C. After overnight incubation, the samples were centrifuged for 10 min at maximum speed (14,000 ×
The quality and concentration of DNA were checked using a NanoDrop apparatus (Thermo Fisher Scientific, Massachusetts, USA). Real-time quantitative PCR (RT-qPCR) was performed with 1.5 µg of DNA and using a Luna Universal qPCR Master Mix (New England Biolabs, Massachusetts, USA) in a StepOnePlus apparatus (Applied Biosystems, Foster City, CA, USA) with specific fungal oligonucleotides (TEF1a). Previously, oligonucleotide efficiency was calculated for each strain and used with the charge control sample Ct (adhesion 100%) to determine the sample adhesion percentage.
IL-8 production by HT29 cells after coculture with fungi
As part of the stimulation protocol, before HT29 stimulation, the cells were stressed for 24 h in medium containing only 5% FBS and not 10%. For stimulation, TNF-α (catalog no. 30001A, Peprotech, France) was added at 5 ng/mL. Coincubation with fungi was realized by adding a yeast suspension with a multiplicity of infection (MOI) of 5 in triplicate, and the plates were incubated for 6 h at 37°C and 5% CO2. Independent experiments were repeated two to four times depending on the sample tested.
After treatment, the culture supernatants were collected for the assay. IL-8 quantification was performed using an IL-8 enzyme-linked immunosorbent assay kit (ELISA kit: ELISA Max Deluxe set catalog no. 431504, BioLegend, France) and following the protocol of the provider. Absorbance was measured at 450 nm using a plate spectrophotometer (Tecan Infinite 200Pro, Switzerland).
Human peripheral blood mononuclear cells (PBMCs) and fungal coincubation
Human PBMCs were purified from whole blood of 27 donors (Etablissement Français du Sang, Le Chesnay, France) using Histopaque-1077 (Sigma Aldrich, France) gradient centrifugation. PBMCs were harvested from the interface, washed three times with sterile PBS, and diluted in 5 mL (for 10 mL of blood) of RPMI 1640 containing 10% FBS (Eurobio Scientific) and 1% penicillin/streptomycin (Sigma-Aldrich, France). The PBMC concentration was determined by using a Kova slide and adjusted to a final concentration of 1 × 106 /mL.
One hundred microliters at 1 × 106 /mL of freshly isolated PBMCs (1 × 106 /mL) were seeded into 96-well culture plates (U-bottom), and 100 µL of a fungal suspension (5 × 106 /mL) was added to each well (triplicate) for a final ratio of 1 (PMBCs):5 (fungi). Lipopolysaccharide (Invitrogen, France) at a final concentration of 10 ng/mL was used as a positive control treatment. To determine cytokine expression, samples were incubated for 24 h at 37°C and 5% CO2, and the supernatant was collected for ELISAs.
Lamina propria immune cell characterization
Cell preparation and stimulation
For isolation of lamina propria immune cells, colons were cut out and open longitudinally. Tissues were washed with cold PBS 1× to remove the fecal and then were cut cross-sectionally into 0.5- to 1-cm-long pieces and then mixed with 5 mL pre-warmed HBSS 1× with 5% fetal calf serum (FCS), EDTA (5 mM, Sigma-Aldrich), and DTT (0.145 mg/mL, Sigma-Aldrich) in the shaker at 37°C for 20 min. The supernatants were discarded, and pellets were washed with cold PBS 1×. The tissue pieces were then transferred to a new tube and digested with collagenase type VIII (0.5 mg/mL, Sigma-Aldrich) supplemented with DNase I (1 mg/mL, Roche) for 30 min. All the contents were passed through a 100-mm cell strainer. Lamina propria lymphocytes (LPLs) were obtained using the 40/80 Percoll centrifugation (GE Healthcare). Cells were centrifuged briefly and suspended in complete RPMI containing 10% (vol/vol) FCS (Eurobio Scientific), 1% HEPES (Gibco), 100 U/mL penicillin, and 100 mg/mL streptomycin (Sigma-Aldrich), and 50 µM 2-mercaptoethanol (Sigma-Aldrich).
For mouse mesenteric lymph node preparation, lymph nodes were homogenized and washed in complete RPMI. Mouse cells were then stimulated in culture medium containing 50 ng/mL PMA (Sigma-Aldrich) and 1 mg/mL ionomycin (Sigma-Aldrich) for 3 h at 37°C, 5% CO2, in the presence of 10 mg/mL Brefeldin-A (Sigma-Aldrich).
Flow cytometry
Single-cell suspensions were prepared in FACS buffer: PBS 1× (Gibco) supplemented with 2% (vol/vol) FCS (Eurobio Scientific), 0.01% (vol/vol) sodium azide (Sigma-Aldrich). Cells were stained on ice in PBS 1× (Gibco) with Zombie Aqua Fixable Viability Kit (BioLegend). Cells were surface stained in FACS buffer with the following antibodies from eBioscience: FITC-labeled anti-mouse CD3ε (145-2C11), anti-CD16/32 (93); from BioLegend: BV785-labeled anti-mouse CD4 (RM4-5) and BV605-labeled anti-mouse CD8a (53-6.7). Cells were washed in FACS buffer. In mouse, CD4+ alpha beta T cells are CD3+ CD4+ CD8−.
After surface staining, cells were fixed using 4% (vol/vol) paraformaldehyde (PFA; Electron Microscopy Sciences) in PBS 1×. Cells were permeabilized in FACS buffer supplemented with 0.5% (wt/vol) saponin (Sigma-Aldrich). Intracellular staining was performed in the same permeabilization buffer with the following antibodies from eBioscience: APC/Cy7-labeled anti-IFN-gamma (XMG1.2); from BioLegend: PE-Cy7 anti-mouse IL-10 (JES5-16E3).
Flow cytometry was carried out by using LSR Fortessa X-20 (BD). Data were analyzed by using FlowJo software (BD Biosciences).
Colitis model in mice
Eight-week-old female C57BL/6J mice were purchased from Janvier Laboratory (Le Genest, France) and used 1 week after reception. Animals were kept in humidity- and temperature-controlled rooms under a 12-h light-dark cycle and had access to a chow diet and water
Prior to DSS administration, the mice were gavaged with a suspension of fungi, 1.107 CFU per gavage/mouse/day. When cyclosporin was mentioned, cyclosporin A (Sigma-Aldrich, France) was administered intraperitoneally as follows: 100 µL per mouse at a concentration of 25 mg/kg, three times per week during the length of the experiments after the beginning of the DSS treatment (9).
One week after starting the fungal administration, mice were given 2% (wt/vol) DSS colitis grade (molecular weight, 36,000–50,000; MP Biomedicals, Solon, OH) dissolved in the drinking water
For quantification of yeast in fresh stool, stool samples were collected over the course of the study to determine the quantity of yeast remaining after intragastric gavage. Fresh stool samples were weighed and suspended in PBS (Gibco Fischer-Scientific, France) in a proportion of 40 µL/mg feces. For quantification, the dilutions of feces were plated on YEPD agar (Sigma-Aldrich France) plates supplemented with ampicillin (100 mg/mL Sigma-Aldrich France) and penicillin/streptomycin (50 mg/mL Gibco Fisher-Scientific France) and incubated at 30°C for 24 to 48 h. The fungi were then counted, and the absolute quantities of yeast were determined according to the corresponding dilutions.
Tissues and samples
Mice were euthanized by cervical dislocation. The distal colon was fixed in 4% paraformaldehyde (Electron Microscopy Sciences, Hatfield, PA, USA), and the proximal colon was flushed and frozen for further RNA extraction. Fecal samples were collected at day 7 and at the end of the protocol (day 12) and frozen for gut microbiota analysis and fecal lipocalin-2 (LCN2) level measurements. All samples were stored at −80°C until use.
Quantification of fecal LCN2 levels
Frozen fecal samples were weighed and suspended in cold PBS. The samples were then agitated on a Precellys (Bertin Corp., France) for 40 s at 5,000 rpm with 4.5 mm glass beads to obtain a homogenous fecal suspension. The samples were then centrifuged for 5 min at 10,000 ×
RNA extraction and gene expression analysis using nanoString technology
Total RNA was isolated from colon samples using a RNeasy Mini Kit (Qiagen, Hilden, Germany), including a DNAse treatment step, according to the manufacturer’s instructions. The quality and concentration of RNA were checked using a NanoDrop apparatus (Thermo Fisher Scientific, Massachusetts, USA). NanoString analysis was performed and analyzed according to the manufacturer’s recommendations. The panel used for this analysis was the “Immunology V1” panel from nanoString; it was composed of 561 genes and described the following pathways: adaptative and innate immune system, apoptosis, autophagy, signaling pathways (B-cell receptor, T cell receptor, NH-kappa B, NLR, TGF-beta, TGF-beta, TLR, TNF family, type I and II interferon, chemokine, and cytokine), cell adhesion, complement system, hemostasis, host-pathogen interaction, immunometabolism, inflammasomes, lymphocyte activation, and trafficking, MHC class I and II antigen presentation, oxidative stress, phagocytosis and degradation, TH1, TH2, Treg, and TH17 differentiation, and transcriptional regulation.
16S DNA gene and ITS2 sequencing
Bacterial diversity was determined for each sample by targeting a portion of the ribosomal genes. PCR was performed to prepare amplicons using V3-V4 oligonucleotides (PCR1F_460: 5′
A similar approach was used for fungal microbiota using the primers ITS2 (sense) 5′-GTGARTCATCGAATCTTT-3′ and (antisense) 5′-GAT
16S and ITS2 sequence analysis
The 16S sequences were demultiplexed and quality filtered using the QIIME version 2.1.0 software package. The sequences were then assigned to OTUs using the UCLUST algorithm with a 97% pairwise identity threshold and classified taxonomically using the SILVA reference database (version 13.8) for bacteria. For the ITS sequences, data were processed using the FROGS pipeline (10) for sequence quality control, and filtering and affiliation of taxa were performed with the UNITE ITS database (version 8_2) (11) using the FROGS guidelines for ITS data (http://frogs.toulouse.inra.fr/). Rarefaction analysis was performed and used to compare the relative abundance of OTUs across samples. Alpha diversity was estimated using the Shannon diversity index or the number of observed species. Beta diversity was measured using the Jaccard distance matrix and was used to build principal coordinates analysis plots. The linear discriminant analysis (LDA) effect size (LEfSe) algorithm was used to identify taxa that were specific to treatment. Deposition of the raw sequence data in the SRA database from the NCBI; the accession numbers are as follows: PRJNA879435.
Statistical analysis
GraphPad Prism version 7 (San Diego, CA, USA) was used for all analyses and preparation of graphs. For all data displayed in graphs, the results are expressed as the mean ± SEM (
RESULTS
Five yeast strains from International Flavors and Fragrances’ (IFF) collection (DGCC), used in food industries for decades, have been selected to test them in a complex model of gut inflammation. The rationale was to directly monitor the potential effects of these strains in a model of inflammation as a screening tool for possible probiotic properties. Some probiotic strains have the capacity to reduce the level of inflammation in this model or to increase the rapidity of recovery. We thus tested the effect of these fungal yeast cells on this model by pretreating the mice with a daily gavage of one strain of yeast for 1 week before beginning the DSS treatment and throughout all DSS administration (Fig. 1A). When treated with 107 yeasts per day of
Fig 1
Dextran sodium sulfate-induced colitis is improved with
However, treatment with
Neither the medium supernatant nor the dead yeast cells have a comparable positive effect on gut inflammation
To identify possible effectors of the two potential probiotics, we tested whether the culture supernatant or the dead cells could elicit comparable protective effects during colitis. Using the same DSS-induced colitis model in mice, both hypotheses were tested. In Fig. S2A and B, pure filtered 24-h spent culture medium of each yeast was given to mice 7 days before and during DSS colitis and compared to fresh medium (Vehicle, YEPD). None of the treatments improved colitis recovery, and in contrast, they had a tendency to worsen the symptoms. For the second experiment, yeast cells were heat killed, washed, and resuspended in PBS before gavage into mice 7 days before and during DSS colitis. Treatment of mice with these samples had no beneficial effect on mouse weight loss independent of the yeast used for the culture (Fig. S2C and D). These results suggest that the two living yeasts are mandatory for the effect on the host and that at least the molecules produced by both yeasts in this rich medium are not directly involved in the effect on colitis recovery.
The persistence capacities of
Since live cells are necessary for the effect on inflammation, we monitored the level of living yeast present in the feces during DSS-induced colitis. This experiment showed that no live
Fig 2
Persistence capacities of
To determine how long
While we have shown that only
As
The persistence of
As a large quantity of live
Fig 3
At this point of the study, no mechanistic explanations could be proposed to elucidate the protective effects of both strains. One possible explanation was a modulation of the host response by the yeast cells. To explore this, we performed several
As surface cells, HT29 epithelial cells have the capacity to produce different types of cytokines to communicate with immune cells at the periphery and regulate the immune system response of the host. One classical proinflammatory molecule produced by HT29 cells is IL-8, which is produced after TNF-α induction. When coincubated individually with the two yeast strains with TNF-α-treated HT29 cells for 6 h, we showed that no significant difference could be observed at an MOI of 1. Nevertheless, at an MOI of 5,
Fig 4
Within the gut, microorganisms also interact with immune cells via the cells present in the lamina propria. Defining how the yeast cells are recognized and what the response elicited when they encounter immune cells can also reveal why these yeasts affect DSS-induced colitis. Hence, PBMCs extracted from the blood of healthy subjects were coincubated with yeast cells at an MOI of 5, and the culture supernatant was subjected to ELISA analyses for cytokine quantification. In Fig. 4B, the ratio of TNF-α to IL-10 production illustrates the level of the proinflammatory effect of each strain, with the use of
Since all yeasts’ effects observed were localized in the gastro-intestinal tract, we also wanted to characterize the effect of both yeast on the local immune cells on naive mice without any inflammation. With this aim, we gavaged the mice for 14 days with either the vehicle,
Colon cytokine profiles after DSS treatment with
The measurement of lipocalin-2 by ELISA, a marker of gut inflammation present in the feces, showed a clear tendency of reduction with
Fig 5
Inflammatory response to
To determine whether this global effect was due directly to the presence of
Microbiota analysis shows a strong impact of
To test whether the effect on inflammation might be indirect and through modulation of the gut microbiota, we performed an amplicon-based analysis of both fungal (ITS2) and bacterial (16S) microbiota of mice after 1 week of gavage with
Fig 6
Bacterial microbiota analysis showed comparable consequences of the gavage of the two yeasts, with no effect of
Fig 7
The bacterial microbiota is significantly modified by
DISCUSSION
Microbiota analysis has reached a considerable level of interest since the 2000s, first through academic research and then quickly in private companies and, more recently, in the public domain. This interest was driven by the discovery of the strong impact the different microbiota can have on human health. The bacterial gut microbiota quickly concentrated the strongest effort due to the higher number of scientists working on bacteria compared to yeasts and to several major discoveries related to human diseases such as diabetes, obesity, and inflammatory bowel diseases (12, 13). Very little interest first was dedicated to the fungal part of the gut microbiota, but with the improvement of the databases and the techniques, the scientific community was able to also link fungal dysbiotic conditions to some human diseases, such as gut inflammation or cancer (14, 15). Interestingly, unlike bacteria, the gut fungal community is largely derived from the food ingested. As such, the fungi used in food processes are central for our comprehension of fungal community modulation and consequently its role.
While the fungi used in food processes are in many cases killed during the process of production, there are foods that are composed of live cells, such as cheeses and some fermented foods or beverages or even some food products that are artificially covered by a chosen microbial community to protect the product from the development of deleterious or pathogenic microbial strains (16). These fungal strains have been used for decades or more in food production than in the food industry and are considered innocuous. However, very little is known about the potential positive effects of these strains on our health, especially now that our techniques of investigation have largely improved and can follow parameters never studied to date. To address this question, we selected five yeast strains from the IFF collection to test their potential probiotic effects on gut health in the context of gut inflammation using
To understand how both yeast strains affected gut sensitivity to inflammation, we investigated their impact on the host inflammatory response with different experiments. One strategy was to detect the anti-inflammatory potential of the strains using an
The use of dead cells or the culture supernatant in the same DSS-induced colitis model did not allow us to identify any effectors of this positive effect on gut inflammation, since none of these fractions had a similar effect. Thus, live yeast cells seem mandatory to observe any effect on the mouse physiopathology of gut inflammation in the context of DSS-induced colitis.
To further monitor the effect of the two yeasts on the mice and to investigate the potential mechanism of action, we determined the modification of the bacterial and fungal microbiota after administration of
This characterization of yeast used in the food industry on their potential probiotic properties allowed for the identification of two strains,
While we cannot describe the complete mechanism at play yet, we can hypothesize that a direct role on the immune pathway with Fkbp and IL-8 is a possibility as well as the modulation of the bacterial and fungal microbiota that would indirectly affect the host susceptibility to gut inflammation with the enrichment in the
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2023 Hugot et al. This work is published under https://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
ABSTRACT
Many strains have been used and selected by the food industry for their capacities to ferment, produce flavors, or produce heterologous molecules. Very little is known about the diversity of foodborne yeasts and their potential effect on gut microbiota and gut health. We initiated a complete characterization of five strains belonging to five species with a long history of safe use in food:
IMPORTANCE
The food industry has always used many strains of microorganisms including fungi in their production processes. These strains have been widely characterized for their biotechnological value, but we still know very little about their interaction capacities with the host at a time when the intestinal microbiota is at the center of many pathologies. In this study, we characterized five yeast strains from food production which allowed us to identify two new strains with high probiotic potential and beneficial effects in a model of intestinal inflammation.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer