Articles Service
Research article
Anti-Inflammatory Response in TNFα/IFNγ-Induced HaCaT Keratinocytes and Probiotic Properties of Lacticaseibacillus rhamnosus MG4644, Lacticaseibacillus paracasei MG4693, and Lactococcus lactis MG5474
Mediogen Co., Ltd., Jecheon 27159, Republic of Korea
Correspondence to:J. Microbiol. Biotechnol. 2023; 33(8): 1039-1049
Published August 28, 2023 https://doi.org/10.4014/jmb.2301.01028
Copyright © The Korean Society for Microbiology and Biotechnology.
Abstract
Keywords
Graphical Abstract
Introduction
Atopic dermatitis (AD) is a common and chronic, relapsing inflammatory skin disease with itchy, pruritic lesions [1]. In particular, since the symptoms of AD appear in visible places such as the hands and ears, they reduce the patient's quality of life and seriously affect their mental health [2]. Over the past decade, AD has markedly increased in prevalence in children and adults, and the resulting economic burden of managing this disease is reported to be significant [3, 4]. Synthetic drugs approved by the FDA to treat AD have been developed, but they cause side effects, such as conjunctivitis and headache [5]. Therefore, for the treatment of AD, preventive agents using natural materials that are safe and free of side effects are urgently needed.
The epidermis of the skin, which is mainly composed of keratinocytes, is the outermost part of the human body and acts as a physical barrier against various triggers [6]. Keratinocytes primarily modulate the immune system in the skin [7]. Stimuli that cause inflammation in keratinocytes include ultraviolet B (UVB), interleukin (IL)‐1α, lipopolysaccharide (LPS), tumor necrosis factor alpha (TNF-α), and interferon gamma (IFN-γ) [8]. Keratinocytes produce chemokines (macrophage-derived chemokine (MDC), thymus- and activation-regulated chemokine (TARC)) and inflammatory cytokines (thymic stromal lymphopoietin (TSLP), IL-4, IL-25, and IL-33), which contribute to an inflammatory response in the skin [9]. When hyper‐immunity occurs in keratinocytes due to harmful triggers, chronic inflammation is induced, which causes atopic dermatitis (AD) accompanied by severe itching. Thus, an immune imbalance in keratinocytes has been implicated in AD pathogenesis [10]. A number of natural products have been reported for their anti-inflammatory efficacy related to atopic‐like responses; however, studies on lactic acid bacteria (LAB) in keratinocytes are still lacking [11].
LAB are a group of gram-positive bacteria, including
Therefore, in this study we investigated the effect of SL on the production of chemokine, including TARC and MDC, and the mRNA expression of IL4, IL‐5, IL‐25, and IL‐33 in TNF-α/IFN-γ-induced HaCaT keratinocytes. We also evaluated the effect of SL on the NF-κB and MAPK signaling pathways in these keratinocytes. Finally, we analyzed the SCFAs and examined the probiotic properties, stability, and safety of the strains with anti-inflammatory activity.
Materials and Methods
Preparation of LAB Strains
All of the LAB strains used in this study were provided by Mediogen Co., Ltd. (Korea) and identified using 16S rRNA gene sequencing (SolGent Co., Ltd., Korea). Isolates were originated from human and/or fermented foods, and the details with NCBI accession numbers are listed in Table 1. LAB were cultured in De Man, Rogosa, and Sharpe (MRS) broth (BD Biosciences, USA) in an anaerobic chamber at 37°C. After 18 h, the SL was collected via centrifugation (4,000 ×
-
Table 1 . The isolates and accession numbers of LAB strains used in this study.
LAB Accession No. Human L. rhamnosus MG4288MW947157.1 L. rhamnosus MG4643ON668169.1 L. rhamnosus MG4644ON668170.1 L. paracasei MG4693OP077096.1 Food Lc. lactis MG5474ON619520.1 Lc. lactis MG5604ON705160.1 L. reuteri MG5462ON619508.1
Cell Culture
HaCaT keratinocytes (provided from the School of Cosmetic Science and Beauty Biotechnology, Semyung University, Korea) and HT‐29 cells (Korea Cell Line Bank, Korea) were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Gibco, USA) containing 10% heat-inactivated fetal bovine serum (FBS; Gibco) and 1%penicillin and streptomycin (P/S; Gibco) [6]. The cells were maintained at 37°C under a 5% CO2 incubator and split every 2–3 days.
Cell Viability
Cell viability was determined using the 3‐(4,5‐dimethyl‐2‐thiazolyl)‐2,5‐diphenyletrazolium bromide (MTT) assay [18]. HaCaT keratinocytes were seeded at a density of 1.5 × 104 cells/well in 96‐well plates. SL from LAB was pre-treated for 1 h and then co-treated with or without TNF-α/IFN-γ (10 ng/ml, each) for 24 h. After removing the culture media, 0.25 mg/ml of MTT solution (Sigma-Aldrich, USA) was added to each well for 2 h. The formazan crystals formed by the MTT solution were dissolved in DMSO (150 μl). Absorbance at 550 nm was measured using a microplate spectrophotometer (Biotek, USA).
Enzyme‐Linked Immunosorbent Assays (ELISA)
The levels of CCL17/TARC and CCL22/MDC were quantified using the Human CCL17/TARC Human and CCL22/MDC DuoSet ELISA Kit (R&D Systems, USA) according to the manufacturer’s instructions. The optical density at 450 nm was measured using a microplate spectrophotometer (Biotek).
Preparation of mRNA and Real‐Time Polymerase Chain Reaction (PCR)
The mRNA was isolated using NucleoZol (MACHEREY‐NAGEL, Gutenberg, Hoerdt Cedex, France) following the manufacturer’s protocol. The cDNA was prepared from the isolated mRNA using the Maxime RT PreMix (iNtRON, Korea). The mRNA expression was analyzed using the CFX96 System (Bio‐Rad, USA) with the AmfiSure qGreen Q‐PCR Master Mix (Gendepot, USA) and the following primers: IL‐4 (forward; f): 5'‐AACAGCCTCACAGAGCAGAAGAC‐3', IL‐4 (reverse; r): 5'‐GTGTTCTTGGAGGCAGCAAAG‐3'; IL‐5 (f): 5'‐TCTACTCATCGAACTCTGCTGA‐3', IL‐5 (r): 5'‐CCCTTGCACAGTTTGACTCTC‐3'; IL‐25 (f): 5'‐TTGTTTGTTTACTCATCACTCAG‐3', IL‐25 (r): 5'‐TCCTCCTCAGAATCATCCA‐3'; and IL‐33 (f): 5'‐TGTCACATTGGGCAAAGTT‐3', IL‐33 (r): 5'‐CAGTAAGCAGTGTTATCAGGAA‐3'. The mRNA expression was normalized using GAPDH (f): 5'‐ACCCACTCCTCCACCTTTG‐3' and GAPDH (r): 5'-ACCCACTCCTCC ACCTTTG‐3'. Relative quantitative expression was analyzed using the 2‐ΔΔCT method and determined as a fold change of the TNF-α/IFN-γ-treated control [19].
Protein Extraction and Western Blotting
Protein lysates were obtained using radioimmunoprecipitation assay (RIPA) cell lysis buffer (Gendepot) containing phosphatase and protease inhibitors (Gendepot). The extracted proteins were quantified at 1 μg/μl using Bradford (Coomassie) reagent (Gendepot). The protein samples were prepared in a 5X sample buffer (iNtron) and heated at 85°C for 10 min. Western blotting was performed using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS‐PAGE) [20]. The samples were loaded onto 10% Tris–glycine gels and electrophoresed in Tris/Glycine/SDS buffer (Bio-Rad). The separated proteins were transferred to polyvinylidene difluoride (PVDF) membranes (Millipore, USA) and washed with TBS-Tween buffer (TBS-T, Gendepot). After blocking with the Smart‐Block 5 min‐Fast Blocking Buffer (Biomax, Republic of Korea), the membranes were incubated with primary antibodies, phosphorylated (p)‐IκB, IκB, p‐nuclear factor kappa B (NF‐κB) p65, extracellular signal‐regulated kinases (ERK), p‐ERK, p‐c‐Jun N‐terminal kinase (JNK), p38, and p‐p38 (Cell Signaling Technology, USA), and NF-κB p65, JNK, and β‐actin (USA), diluted to 1:1000 overnight at 4°C. After washing with TBS‐T, membranes were incubated with HRP‐conjugated secondary antibodies (1:5000, Gendepot) for 1 h. The western blot images were developed using the LuminoGraph III Lite (Atto Corp., Japan) with the EzWest Lumi Plus (Atto), and density graph analysis was performed using the CS Analyzer 4 (Atto).
Gas Chromatograpy/Mass Spectrometry (GC/MS) Analysis of SCFAs
SCFAs in SL were analyzed using GC–MS (QP2020 NXW/ORP230, Shimadzu, Japan) equipped with a Stabilwax-DA column (60 m × 0.32 mm × 0.25 μm, Shimadzu) according to a previous study [21]. GC analysis was conducted in splitless mode using helium (flow rate, 2 ml/min) as the carrier gas. The oven temperature was held at 50°C for 2 min and then ramped up to 200°C at 10°C/min. This temperature was held for 5 min. The ionization was operated using the following settings: electron energy = 70 eV, ion source heater temperature = 200°C and 150°C, and scan speed = 0.2 s/scan. The analytes were monitored in the selected ion monitor (SIM) mode. Acetic acid, propionic acid, and butyric acid (Sigma-Aldrich) were used as standards and the quantitative analysis in CFS was calculated using the standard curve.
Profiling of Carbohydrate Fermentation and Enzymatic Activity
Carbohydrate fermentation of LAB was measured using the API 50 CHL Kit (BioMerieux, France) [22]. Briefly, LAB strains were grown on MRS agar (BD Biosciences) plates for 18 h, and the colonies were adjusted in API suspension medium (BioMerieux) to 2.0 McFarland. The suspended LAB strains were inoculated into strips and incubated at 37°C for 48 h. Meanwhile, the enzymatic activities of LAB were confirmed using the API ZYM Enzyme System (BioMerieux) [23]. After streaking on MRS medium, a colony was cultured at 37°C for 18 h, and the turbidity was adjusted to 5.0–6.0 McFarland in API suspension medium (BioMerieux). The suspension medium was dispensed on each enzyme of the API ZYM strip and incubated at 37°C for 4 h. The results were confirmed by the color changes.
Cytotoxicity to HT‐29 Cells
Cytotoxicity was estimated using the MTT assay [17]. HT‐29 cells were seeded at a density of 2.5 × 104 cells/well in 96‐well plates. LAB were treated at 106, 107, and 108 CFU/ml for 24 h, and then the culture medium was removed and MTT solution (0.25 mg/ml) was added to each cell for 2 h. The formazan crystals, formed by MTT solution, were dissolved in DMSO (150 μl). Absorbance at 550 nm was measured using a microplate spectrophotometer (Biotek).
Hemolytic and Bile Salt Hydrolase (BSH) Activity
Hemolytic activity was determined using Columbia agar (Oxoid, UK) containing 5% sheep blood (MBcell, Korea) [24]. LAB strains were streaked and cultured at 37°C for 48 h. Hemolysis was observed as halo zones around the colony: (1) alpha hemolysis appeared green in the medium, suggesting partial hemolysis; (2) beta hemolysis showed a clear zone, indicating destroyed red blood cells; and (3) gamma hemolysis did not show any reaction in the surrounding medium, indicating a lack of hemolysis [25]. Meanwhile, BSH enzyme activity of the LAB strains was determined according to a previous report [26]. Briefly, each strain was cultured on MRS agar containing 0.5% taurodeoxycholic acid hydrate (Sigma-Aldrich) and incubated at 37°C under anaerobic conditions for 48 h. BSH activity was assessed with the appearance of a precipitation zone surrounding the colony.
Survival in Gastrointesinal Tract (GIT) Conditions
Survival under conditions similar to those in the GIT environment was assessed as previously described [27]. The viability of LAB was measured after 2 h in PBS (pH 2.5) with 0.3% pepsin (Sigma-Aldrich) as gastric fluids at 37°C, and after 2.5 h in PBS (pH 7.4) with 1% pancreatin‐bile salt (Sigma-Aldrich) as intestinal fluids at 37°C. The live colony count was performed using an MRS agar plate.
Adhesion to HT‐29 Cells
The adhesion test to HT‐29 cells, a human colon adenocarcinoma cell line, was performed as reported previously with some modifications [28]. Briefly, HT‐29 cells were sub‐cultured at 1.5 × 105 cells/well in 12‐well plates and grown at 37°C in a humidified atmosphere of 5% CO2 until they formed a monolayer. The cells were then treated with LAB (1 × 108 CFU/ml) for 4 h. After treatment, the HT‐29 cells were washed three times with PBS and then lysed with 1 ml of PBS. The cell lysate was diluted with buffered peptone water (Oxoid), plated on MRS agar, and incubated at 37°C for 48 h. Adhesion ability (%) was calculated using the following equation: log (adherent counts of LAB) CFU/ml/log (initial counts of LAB) CFU/ml × 100.
Statistical Analysis
Results are expressed as mean ± standard error of the mean (SEM). Statistical significance was analyzed using one-way analysis of variance (ANOVA) followed by post hoc verification with Duncan’s multiple comparison tests (SPSS Statistics ver. 21, IBM, USA).
Results
SL Suppressed Chemokines in TNF-α/ IFN-γ‐Induced HaCaT Keratinocytes
HaCaT keratinocytes were treated with various concentrations of SL to optimize the concentration. The results showed no toxicity at 5% SL (Fig. 1A). As shown in Fig. 1B, 5% SL was not cytotoxic to TNF-α/IFN-γ‐induced HaCaT keratinocytes; therefore, further experiments were conducted at this concentration. TNF-α/IFN-γ-treated HaCaT keratinocytes showed significantly upregulated TARC levels (Fig. 1C). Conversely, treatment with SL from LAB and TNF-α/IFN-γ markedly decreased TARC levels (
-
Fig. 1. Effect of SL (5%) from LAB on cell viability (A), expression of TARC (B) and MDC (C) in TNF-α/IFN- γ‐induced HaCaT keratinocytes.
The bars indicate the mean ± SEM. Different letters (a–f) show significant difference at
p < 0.05 (n = 3). *T+I, TNF-α/IFN-γ only treated group.
Inhibition of Cytokines Related to Immune Response by SL in TNF-α/IFN-γ‐Induced HaCaT Keratinocytes
To determine the effect of different SL on cytokines, IL‐4, IL‐5 , IL‐25, IL‐33, and thymic stromal lymphopoietin (TSLP) were analyzed using real-time PCR and ELISA. As shown in Fig. 2A, IL‐4 and IL‐5 mRNA levels were significantly elevated in TNF-α/IFNγ-induced HaCaT keratinocytes (
-
Fig. 2. Effect of SL (5%) from LAB strains on
IL‐4 ,IL‐5 mRNA expression (A), TSLP (B) and IL‐25, IL‐33 mRNA expression (C) in TNF-α/IFN-γ‐induced HaCaT keratinocytes. The bars indicate the mean ± SEM. Different letters (a–e) show significant difference atp < 0.05 (n = 3). *T+I, TNF-α/IFN-γ only treated group.
Effect of SL on NF‐κB/MAPK Signaling Pathways in TNF-α/ IFN-γ‐Induced HaCaT Keratinocytes
To investigate the effect of SL on the NF-κB signaling pathway, p‐IκB and p‐NF‐κB were analyzed using western blotting (Fig. 3). Increased p‐IκB and p‐NF‐κB levels were observed in TNF-α/IFN-γ-induced HaCaT keratinocytes (
-
Fig. 3. Effect of SL (5%) from LAB strains on the phosphorylation of nuclear factor kappa B (Nf‐κB) signaling pathway in TNF-α/IFN-γ‐induced HaCaT keratinocytes.
The protein expression of IκB (A) and Nf‐κB (B) were normalized to non‐phosphorylated protein. The bars indicate the mean ± SEM. Different letters (a–d) show significant difference at
p < 0.05 (n = 3). *T+I, TNF-α/IFN-γ only treated group.
-
Fig. 4. Modulation of the mitogen‐activated protein kinase (MAPK) signaling by SL (5%) from LAB strains in TNF-α/IFN-γ‐induced HaCaT keratinocytes.
Representative western blot images of p‐ERK/ERK (A), p‐JNK/JNK (B), and p‐p38/p38 (C) expression was shown. The protein expression was normalized to non‐phosphorylated protein. The bars indicate the mean ± SEM. Different letters (a–d) show significant difference at
p < 0.05 (n = 3). *T+I, TNF-α/IFN-γ only treated group.
Analysis of SCFAs in SL from L. rhamnosus MG4644, L. paracasei MG4693, and Lc. lactis MG5474
To analyze SCFAs, which are major metaboiltes of probiotics produced by each strain, the contents of acetic, propionic and butyric acid in SL were measured using GC-MS (Table 2). The calibration curve formulas of acetic, propionic and butyric acid were confirmed as y = 35590x + 44682, y = 105472x + 10648, and y = 241306x + 3248.1, respectively, and through this, quantitative analysis of SCFA in SL was performed. All three strains were confirmed to have high levels of acetic acid content; however,
-
Table 2 . SCFA contents in CFS of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474.LAB SCFAs (mg/l) Acetic acid Propionic acid Butyric acid Total MG4644 2987.00 ± 65.79ab 1.49 ± 0.06a 5.37 ± 0.04a 2993.85 ± 65.69ab MG4693 3112.43 ± 79.35a 1.29 ± 0.11a 4.83 ± 0.22a 3118.55 ± 79.68a MG5474 2643.22 ± 136.40b 1.44 ± 0.03a 4.97 ± 0.19a 2649.63 ± 136.63b Data are presented as mean ± SEM. Different letters (a–d) show significant difference at
p < 0.05 (n = 3).
Probiotic Properties of L. rhamnosus MG4644, L. paracasei MG4693, and Lc. lactis MG5474
As shown in the carbohydrate fermentation profiles, all three strains fermented various sugars (Table 3). We found that all strains utilized L-arabinose, methyl-β-D-xyloside, D-galactose, D-glucose, D-fructose, methyl-α-D-glucoside, arbutin, esculin, salicin, D-cellobiose, D-maltose, D-melibiose, and D-sucrose, and did not utilize glycerol, D-xylose, L-xylose, D-adonitol, D-sorbitol, starch, glycogen, D-turanose, D-tagatose, L-fucose, D-arabitol, gluconate, and 2-keto-gluconate. Each strain differed in its ability to utilize the remaining carbohydrates. Based on these results, the three strains were identified as
-
Table 3 . Carbohydrate fermentation profiles of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474.No. Type of testa MG4644 MG4693 MG5474 No. Type of test MG4644 MG4693 MG5474 1 Control - - - 26 Esculin + + + 2 Glycerol - - - 27 Salicin + + + 3 Erythritol - - - 28 D-Cellobiose + + + 4 D-Arabinose + - - 29 D-Maltose + + + 5 L-Arabinose - - + 30 D-Lactose + + + 6 D-Ribose + + + 31 D-Melibiose - - + 7 D-Xylose - - + 32 D-Sucrose + + + 8 L-Xylose - - - 33 D-Trehalose + + + 9 D-Adonitol - - - 34 Inulin - + - 10 Methyl-β-D-xyloside - - - 35 D-Melezitose + + - 11 D-Galactose + + + 36 D-Raffinose - - + 12 D-Glucose + + + 37 Starch - - + 13 D-Fructose + + + 38 Glycogen - - - 14 D-Mannose + + + 39 Xylitol - - - 15 L-Sorbose + - - 40 Gentiobiose + - + 16 L-Rhamnose + - - 41 D-Turanose + + - 17 Dulcitol + - - 42 D-Lyxose - - - 18 Inositol + - - 43 D-Tagatose + + - 19 D-Mannitol + + - 44 D-Fucose - - - 20 D-Sorbitol + - - 45 L-Fucose + - - 21 Methyl-α-D-mannoside - - - 46 D-Arabitol - - - 22 Methyl-α-D-glucoside + + - 47 L-Arabitol - - - 23 N-Acetylglucosamine + + + 48 Gluconate + + + 24 Amygdalin + - - 49 2-Keto-gluconate - - - 25 Arbutin + - + 50 5-Keto-gluconate - - - aThe mark of the result tests: -, not growth; +, growth.
-
Table 4 . Enzymatic profile of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474.Enzyme assayed fora MG4644 MG4693 MG5474 Control (Negative) 0 0 0 Alkaline phosphatase 1 0 0 Esterase (C4) 3 3 0 Esterase Lipase (C8) 2 3 0 Lipase (C14) 0 0 0 Leucine arylamidase 5 5 5 Valine arylamidase 5 5 2 Crystine arylamidase 2 1 2 Trypsin 0 0 0 α-Chymotrypsin 1 0 1 Acid phosphatase 2 3 3 Naphtol-AS-BI-phosphohydrolase 5 4 2 α-Galactosidase 0 0 0 β-Galactosidase 3 4 0 β-Glucuronidase 0 0 0 α-Glucosidase 2 4 0 β-Glucosidase 5 0 0 N–acetyl -β- glucosaminidase 0 1 0 α-Mannosidase 0 0 0 α-Fucosidase 2 0 0 aData are indicated on scale of 0 (no reaction) to 5 (maximum activity).
Safety of L. rhamnosus MG4644, L. paracasei MG4693, and Lc. lactis MG5474
To confirm the safety of
-
Fig. 5. Hemolysis (A), BSH activity (B), and cytotoxicity of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474 in HT‐29 cells (C). HT‐29 cells were treated withL. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474 (106, 107, and 108 cells/ml). Data are presented as the mean±SEM (n =3), and have no statistical significance.
Stability in Simulated GIT and Adhesion to HT‐29 Cells of L. rhamnosus MG4644, L. paracasei MG4693, and Lc. lactis MG5474
The stability in simulated GIT of
-
Fig. 6. Cytotoxicity in HT‐29 cells (A), hemolysis (B), and BSH activity (C) of
L. rhamnosus MG4644,L. paracasei MG4693, and Lc. lactis MG5474. HT‐29 cells were treated withL. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474 (106, 107, and 108 cells/ml). Data are presented as the mean ± SEM (n = 3), and have no statistical significance.
Discussion
The gut-skin axis is currently being investigated based on studies showing that the immune regulation ability of the gut microbiome affects this distant organ [30]. Studies have reported that the skin and intestines have many similarities, and that skin conditions can be affected by changes in gut microbes [30]. The intestinal microflora acts on AD in two directions: (1) directly by producing metabolites such as SCFAs and delivering them to the skin through the blood vessels, and/or (2) indirectly by controlling the microbiota, which helps balance the immune system [31]. In a clinical study,
The stimulation of TNF-α and IFN-γ results in the expression of many cytokines (IL family and TSLP) and chemokines (MDC and TARC) in keratinocytes [36]. TARC and MDC, which are specific ligands for the CC motif chemokine ligand 4 (CCR4) expressed by Th2 cells, have been reported to correlate with the pathogenesis of AD. In our study, most SL downregulated MDC and TARC levels, which were elevated by TNF-α/IFN-γ in keratinocytes. The incidence of AD-like inflammation can be improved by regulating cytokines involved in T-helper cell type (Th)-2 and Th-1/Th-2 ratio [15]. Inhibition of Th2 cytokines such as IL-4 and IL-5 is known to block the progression of AD-like inflammation [11]. TSLP, which induces Th2 differentiation and produces Th2 cytokines, contributes to pruritus in keratinocytes of patients with AD [10]. In the present study, the mRNA expression of IL-4, IL-5, and TSLP were most inhibited by
Activation of NF-κB and MAPKs signaling pathways related to inflammation in keratinocytes is induced by cross-linking with TNF-α and IFN-γ [9]. NF-κB, stimulated by the degradation of IκB, is a major transcription factor involved in the inflammatory responses in AD [35]. Phosphorylated NF-κB translocates to the nucleus and produces cytokines and chemokines to regulate inflammatory and immune responses [36]. In our results, SL significantly diminished p‐IκB and p‐NF‐κB increased by TNF-α/IFN-γ in keratinocytes. Furthermore, NF-κB is regulated by the MAPK signaling pathway [36]. A previous study reported that inhibition of p‐p38 and p‐ERK attenuates skin inflammation [9]. This study showed that SL significantly inhibited p‐p38 and p‐ERK in TNF-α/IFN-γ-induced HaCaT keratinocytes. In summary,
The FAO/WHO Working Group reported that LAB strains should demonstrate probiotic properties, including tolerance under gastrointestinal tract (GIT) conditions and bile salt hydrolase (BSH) de‐conjugation, adhesion to intestinal epithelial cells, and safety (hemolysis and cytotoxicity) [7]. Safety evaluation of LAB strains is the first priority for their use as probiotics because they are rarely used as causative agents of potential opportunistic infections [41]. The MTT assay can measure mitochondrial activity in living cells and is therefore used to measure the cytotoxic effects of samples in vitro [42]. In addition, this assay is widely used as a cytotoxic screening method to evaluate the safety of probiotics [43]. Our results showed that all three strains were non-toxic to intestinal epithelial cells. In addition, hemolysis and BSH activities are considered important safety traits for probiotics [41]. As bile salts are synthesized from cholesterol, some LAB with BSH activity have been suggested to prevent hypercholesterolemia [44]. However, excessive BSH activity can have detrimental effects on the host by causing lipid dyspepsia and impairing colonic mucosal function, potentially leading to the formation of gallstones [45]. In this study,
For consumption as a probiotic, stability in the GIT in the host is important [13]. In other words, probiotics must tolerate bile salt concentrations, enzymes such as pepsin and pancreatin, and low pH (2.5–3.5) [46]. This study found that
Conclusion
In the current study,
Conflict of Interest
The authors have no financial conflicts of interest to declare.
References
- Torres T, Ferreira E, Gonçalo M, Mendes-Bastos P, Selores M, Filipe P. 2019. Update on atopic dermatitis.
Acta Medica Port. 32 : 606-613. - Yoo J, Koo HYR, Han K, Lee YB. 2022. Impairment of quality of life and mental health status in adult-onset atopic dermatitis.
Ann. Dermatol. 34 : 278-286. - Ha J, Lee SW, Yon DK. 2020. Ten-year trends and prevalence of asthma, allergic rhinitis, and atopic dermatitis among the Korean population, 2008-2017.
Clin. Exp. Pediatr. 63 : 278-283. - Manjelievskaia J, Boytsov N, Brouillette MA, Onyekwere U, Pierce E, Goldblum O,
et al . 2021. The direct and indirect costs of adult atopic dermatitis.J. Manag. Care Spec. Pharm. 27 : 1416-1425. - Albader SS, Alharbi AA, Alenezi RF, Alsaif FM. 2019. Dupilumab side effect in a patient with atopic dermatitis: a case report study.
Biologics 13 : 79-82. - Lee JY, Kim Y, Kim J-I, Lee H-Y, Moon G-S, Kang C-H. 2022. Improvements in human keratinocytes and antimicrobial effect mediated by cell-free supernatants derived from probiotics.
Fermentation 8 : 332. - Bitschar K, Wolz C, Krismer B, Peschel A, Schittek B. 2017. Keratinocytes as sensors and central players in the immune defense against
Staphylococcus aureus in the skin.J. Dermatol. Sci. 87 : 215-220. - Albanesi C, Scarponi C, Giustizieri ML, Girolomoni G. 2005. Keratinocytes in inflammatory skin diseases.
Curr. Drug Targets Inflamm. Allergy 4 : 329-334. - Cho SH, Kim HS, Lee W, Han EJ, Kim SY, Fernando IS,
et al . 2020. Eckol fromEcklonia cava ameliorates TNF-α/IFN-γ-induced inflammatory responses via regulating MAPKs and NF-κB signaling pathway in HaCaT cells.Int. Immunopharmacol. 82 : 106146. - Chieosilapatham P, Kiatsurayanon C, Umehara Y, Trujillo-Paez J, Peng G, Yue H,
et al . 2021. Keratinocytes: Innate immune cells in atopic dermatitis.Clin. Expe. Immunol. 204 : 296-309. - Yan F, Li F, Liu J, Ye S, Zhang Y, Jia J,
et al . 2020. The formulae and biologically active ingredients of Chinese herbal medicines for the treatment of atopic dermatitis.Biomed. Pharmacother. 127 : 110142. - Edalati E, Saneei B, Alizadeh M, Hosseini SS, Bialvaei AZ, Taheri K. 2019. Isolation of probiotic bacteria from raw camel's milk and their antagonistic effects on two bacteria causing food poisoning.
New Microb. New Infect. 27 : 64-68. - Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B,
et al . 2014. The international scientific association for robiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic.Nat. Rev. Gastroenterol. Hepatol. 11 : 506-514. - Lizardo MV, Tavaria FK. 2022. Probiotics and skin health, pp. 389-405. Probiotics, Ed. Elsevier.
- Rusu E, Enache G, Cursaru R, Alexescu A, Radu R, Onila O,
et al . 2019. Prebiotics and probiotics in atopic dermatitis.Exp. Ther. Med. 18 : 926-931. - Lew LC, Liong MT. 2013. Bioactives from probiotics for dermal health: functions and benefits.
J. Appl. Microbiol. 114 : 1241-1253. - Lee JY, Kang JH, Jung YR, Kang CH. 2022.
Lactobacillus gasseri MG4247 andLacticaseibacillus paracasei MG4272 and MG4577 modulate allergic inflammatory response in RAW 264.7 and RBL-2H3 cells.Probiotics Antimicrob. Proteins 31 : 1-10. - Tolosa L, Donato MT, Gómez-Lechón MJ. 2015. General cytotoxicity assessment by means of the MTT assay, pp. 333-348. Protocols in in vitro hepatocyte research, Ed. Springer.
- Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method.
Methods 25 : 402-408. - Bass JJ, Wilkinson DJ, Rankin D, Phillips BE, Szewczyk NJ, Smith K,
et al . 2017. An overview of technical considerations for Western blotting applications to physiological research.Scand. J. Med. Sci. Sports 27 : 4-25. - Park JY, Lee JY, Kim Y, Kang CH. 2023.
Latilactobacillus sakei Wikim0066 protects skin through MMP regulation on UVB-irradiated in vitro and in vivo model.Nutrients 15 : 726. - Syed Yaacob SN, Huyop F, Kamarulzaman Raja Ibrahim R, Wahab RA. 2018. Identification of
Lactobacillus spp. andFructobacillus spp. isolated from fresh Heterotrigona itama honey and their antagonistic activities against clinical pathogenic bacteria.J. Apic. Res. 57 : 395-405. - Kang CH, Kim Y, Han SH, Jeong Y, Park H, Paek NS. 2019. Probiotic properties of bifidobacteria isolated from feces of infants.
J. Milk Sci. Biotechnol. 37 : 40-48. - Yasmin I, Saeed M, Khan WA, Khaliq A, Chughtai MFJ, Iqbal R,
et al . 2020. In vitro probiotic potential and safety evaluation (hemolytic, cytotoxic activity) ofBifidobacterium strains isolated from raw camel milk.Microorganisms 8 : 354. - Buxton R. 2005. Blood agar plates and hemolysis protocols. American Society for Microbiology. pp. 1-9.
- Shehata M, El Sohaimy S, El-Sahn MA, Youssef M. 2016. Screening of isolated potential probiotic lactic acid bacteria for cholesterol lowering property and bile salt hydrolase activity.
Ann. Agric. Sci. 61 : 65-75. - Kim S, Lee JY, Jeong Y, Kang CH. 2022. Antioxidant activity and probiotic properties of lactic acid bacteria.
Fermentation 8 : 29. - Fonseca HC, de Sousa Melo D, Ramos CL, Dias DR, Schwan RF. 2021. Probiotic properties of lactobacilli and their ability to inhibit the adhesion of enteropathogenic bacteria to Caco-2 and HT-29 cells.
Probiotics Antimicrob. Proteins 13 : 102-112. - Cheng KW, Tseng CH, Chen IJ, Huang BC, Liu HJ, Ho KW,
et al . 2022. Inhibition of gut microbial β-glucuronidase effectively prevents carcinogen-induced microbial dysbiosis and intestinal tumorigenesis.Pharmacol. Res. 177 : 106115. - De Pessemier B, Grine L, Debaere M, Maes A, Paetzold B, Callewaert C. 2021. Gut-skin axis: current knowledge of the interrelationship between microbial dysbiosis and skin conditions.
Microorganisms 9 : 353. - Lee SY, Lee E, Park YM, Hong SJ. 2018. Microbiome in the gut-skin axis in atopic dermatitis.
Allergy, Asthma Immunol. Res. 10 : 354-362. - Lolou V, Panayiotidis MI. 2019. Functional role of probiotics and prebiotics on skin health and disease.
Fermentation 5 : 41. - De Vuyst E, Salmon M, Evrard C, Lambert de Rouvroit C, Poumay Y. 2017. Atopic dermatitis studies through in vitro models.
Front. Med. 4 : 119. - Song HK, Park SH, Kim HJ, Jang S, Kim T. 2022.
Spatholobus suberectus dunn water extract ameliorates atopic dermatitis-like symptoms by suppressing proinflammatory chemokine production in vivo and in vitro.Front. Pharmacol. 13 : 919230. - Hwang DH, Koh PO, Kang C, Kim E. 2021. Rosa davurica Pall. improves DNCB-induced atopic dermatitis in mice and regulated TNF-alpha/IFN-gamma-induced skin inflammatory responses in HaCaT cells.
Phytomedicine 91 : 153708. - Yang CC, Hung YL, Ko WC, Tsai YJ, Chang JF, Liang CW,
et al . 2021. Effect of neferine on DNCB-induced atopic dermatitis in HaCaT cells and BALB/c mice.Int. J. Mol. Sci. 22 : 8237. - Sebastian K, Borowski A, Kuepper M, Friedrich K. 2008. Signal transduction around thymic stromal lymphopoietin (TSLP) in atopic asthma.
Cell Commun. Signal. 6 : 5. - Divekar R, Kita H. 2015. Recent advances in epithelium-derived cytokines (IL-33, IL-25 and TSLP) and allergic inflammation.
Curr. Opin. Allergy Clin. Immunol. 15 : 98. - Xiao X, Hu X, Yao J, Cao W, Zou Z, Wang L,
et al . 2022. The role of short-chain fatty acids in inflammatory skin diseases.Front. Microbiol. 13 : 1083432. - Kang CH, Kim JS, Park HM, Kim S, Paek NS. 2021. Antioxidant activity and short-chain fatty acid production of lactic acid bacteria isolated from Korean individuals and fermented foods.
3 Biotech. 11 : 217. - Aryantini NPD, Yamasaki E, Kurazono H, Sujaya IN, Urashima T, Fukuda K. 2017. In vitro safety assessments and antimicrobial activities of
Lactobacillus rhamnosus strains isolated from a fermented marés milk.Anim. Sci. J. 88 : 517-525. - Meerloo JV, Kaspers GJ, Cloos J. 2011. Cell sensitivity assays: the MTT assay, pp. 237-245. Cancer cell culture, Ed. Springer.
- Ranjan KG, Sankar GG, Raju D. 2020. In vitro evaluation of antibacterial, cytotoxic and adherence studies of selected commercial probiotics.
J. Pure Appl. Microbiol. 14 : 2085-2091. - Khare A, Gaur S. 2020. Cholesterol-lowering effects of
Lactobacillus species.Curr. Microbiol. 77 : 638-644. - Byakika S, Mukisa IM, Byaruhanga YB, Muyanja C. 2019. A review of criteria and methods for evaluating the probiotic potential of microorganisms.
Food Rev. Int. 35 : 427-466. - Casarotti SN, Todorov SD, Penna ALB. 2015. Effect of different matrices on probiotic resistance to in vitro simulated gastrointestinal conditions.
Int. J. Dairy Technol. 68 : 595-601. - Jovanović JN, Nikolić B, Šeatović S, Zavišić G, Mitić-Ćulafić D, Vuković-Gačić B,
et al . 2015. Characterization of some potentially probioticLactobacillus strains of human origin.Food Sci. Biotechnol. 24 : 1781-1788. - Verhoeckx K, Cotter P, López-Expósito I, Kleiveland C, Lea T, Mackie A,
et al . 2015. The impact of food bioactives on health: in vitro and ex vivo models.Cham CH : Springer.
Related articles in JMB
Article
Research article
J. Microbiol. Biotechnol. 2023; 33(8): 1039-1049
Published online August 28, 2023 https://doi.org/10.4014/jmb.2301.01028
Copyright © The Korean Society for Microbiology and Biotechnology.
Anti-Inflammatory Response in TNFα/IFNγ-Induced HaCaT Keratinocytes and Probiotic Properties of Lacticaseibacillus rhamnosus MG4644, Lacticaseibacillus paracasei MG4693, and Lactococcus lactis MG5474
Ji Yeon Lee, Jeong‐Yong Park, Yulah Jeong, and Chang‐Ho Kang*
Mediogen Co., Ltd., Jecheon 27159, Republic of Korea
Correspondence to:Chang‐Ho Kang, changho-kang@naver.com
Abstract
Atopic dermatitis (AD) is a chronic inflammatory disease caused by immune dysregulation. Meanwhile, the supernatant of lactic acid bacteria (SL) was recently reported to have anti-inflammatory effects. In addition, HaCaT keratinocytes stimulated by tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ) are widely used for studying AD‐like responses. In this study, we evaluated the anti-inflammatory effects of SL from lactic acid bacteria (LAB) on TNF-α/IFN-γ-induced HaCaT keratinocytes, and then we investigated the strains’ probiotic properties. SL was noncytotoxic and regulated chemokines (macrophage-derived chemokine (MDC) and thymus and activation-regulated chemokine (TARC)) and cytokines (interleukin (IL)‐4, IL‐5, IL‐25, and IL‐33) in TNF-α/IFN-γ-induced HaCaT keratinocytes. SL from Lacticaseibacillus rhamnosus MG4644, Lacticaseibacillus paracasei MG4693, and Lactococcus lactis MG5474 decreased the phosphorylation of nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK). Furthermore, the safety of the three strains was demonstrated via hemolysis, bile salt hydrolase (BSH) activity, and toxicity tests, and the stability was confirmed under simulated gastrointestinal conditions. Therefore, L. rhamnosus MG4644, L. paracasei MG4693, and Lc. lactis MG5474 have potential applications in functional food as they are stable and safe for intestinal epithelial cells and could improve atopic inflammation.
Keywords: Anti‐inflammation, cell‐free supernatant, lactic acid bacteria, probiotics, keratinocytes
Introduction
Atopic dermatitis (AD) is a common and chronic, relapsing inflammatory skin disease with itchy, pruritic lesions [1]. In particular, since the symptoms of AD appear in visible places such as the hands and ears, they reduce the patient's quality of life and seriously affect their mental health [2]. Over the past decade, AD has markedly increased in prevalence in children and adults, and the resulting economic burden of managing this disease is reported to be significant [3, 4]. Synthetic drugs approved by the FDA to treat AD have been developed, but they cause side effects, such as conjunctivitis and headache [5]. Therefore, for the treatment of AD, preventive agents using natural materials that are safe and free of side effects are urgently needed.
The epidermis of the skin, which is mainly composed of keratinocytes, is the outermost part of the human body and acts as a physical barrier against various triggers [6]. Keratinocytes primarily modulate the immune system in the skin [7]. Stimuli that cause inflammation in keratinocytes include ultraviolet B (UVB), interleukin (IL)‐1α, lipopolysaccharide (LPS), tumor necrosis factor alpha (TNF-α), and interferon gamma (IFN-γ) [8]. Keratinocytes produce chemokines (macrophage-derived chemokine (MDC), thymus- and activation-regulated chemokine (TARC)) and inflammatory cytokines (thymic stromal lymphopoietin (TSLP), IL-4, IL-25, and IL-33), which contribute to an inflammatory response in the skin [9]. When hyper‐immunity occurs in keratinocytes due to harmful triggers, chronic inflammation is induced, which causes atopic dermatitis (AD) accompanied by severe itching. Thus, an immune imbalance in keratinocytes has been implicated in AD pathogenesis [10]. A number of natural products have been reported for their anti-inflammatory efficacy related to atopic‐like responses; however, studies on lactic acid bacteria (LAB) in keratinocytes are still lacking [11].
LAB are a group of gram-positive bacteria, including
Therefore, in this study we investigated the effect of SL on the production of chemokine, including TARC and MDC, and the mRNA expression of IL4, IL‐5, IL‐25, and IL‐33 in TNF-α/IFN-γ-induced HaCaT keratinocytes. We also evaluated the effect of SL on the NF-κB and MAPK signaling pathways in these keratinocytes. Finally, we analyzed the SCFAs and examined the probiotic properties, stability, and safety of the strains with anti-inflammatory activity.
Materials and Methods
Preparation of LAB Strains
All of the LAB strains used in this study were provided by Mediogen Co., Ltd. (Korea) and identified using 16S rRNA gene sequencing (SolGent Co., Ltd., Korea). Isolates were originated from human and/or fermented foods, and the details with NCBI accession numbers are listed in Table 1. LAB were cultured in De Man, Rogosa, and Sharpe (MRS) broth (BD Biosciences, USA) in an anaerobic chamber at 37°C. After 18 h, the SL was collected via centrifugation (4,000 ×
-
Table 1 . The isolates and accession numbers of LAB strains used in this study..
LAB Accession No. Human L. rhamnosus MG4288MW947157.1 L. rhamnosus MG4643ON668169.1 L. rhamnosus MG4644ON668170.1 L. paracasei MG4693OP077096.1 Food Lc. lactis MG5474ON619520.1 Lc. lactis MG5604ON705160.1 L. reuteri MG5462ON619508.1
Cell Culture
HaCaT keratinocytes (provided from the School of Cosmetic Science and Beauty Biotechnology, Semyung University, Korea) and HT‐29 cells (Korea Cell Line Bank, Korea) were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Gibco, USA) containing 10% heat-inactivated fetal bovine serum (FBS; Gibco) and 1%penicillin and streptomycin (P/S; Gibco) [6]. The cells were maintained at 37°C under a 5% CO2 incubator and split every 2–3 days.
Cell Viability
Cell viability was determined using the 3‐(4,5‐dimethyl‐2‐thiazolyl)‐2,5‐diphenyletrazolium bromide (MTT) assay [18]. HaCaT keratinocytes were seeded at a density of 1.5 × 104 cells/well in 96‐well plates. SL from LAB was pre-treated for 1 h and then co-treated with or without TNF-α/IFN-γ (10 ng/ml, each) for 24 h. After removing the culture media, 0.25 mg/ml of MTT solution (Sigma-Aldrich, USA) was added to each well for 2 h. The formazan crystals formed by the MTT solution were dissolved in DMSO (150 μl). Absorbance at 550 nm was measured using a microplate spectrophotometer (Biotek, USA).
Enzyme‐Linked Immunosorbent Assays (ELISA)
The levels of CCL17/TARC and CCL22/MDC were quantified using the Human CCL17/TARC Human and CCL22/MDC DuoSet ELISA Kit (R&D Systems, USA) according to the manufacturer’s instructions. The optical density at 450 nm was measured using a microplate spectrophotometer (Biotek).
Preparation of mRNA and Real‐Time Polymerase Chain Reaction (PCR)
The mRNA was isolated using NucleoZol (MACHEREY‐NAGEL, Gutenberg, Hoerdt Cedex, France) following the manufacturer’s protocol. The cDNA was prepared from the isolated mRNA using the Maxime RT PreMix (iNtRON, Korea). The mRNA expression was analyzed using the CFX96 System (Bio‐Rad, USA) with the AmfiSure qGreen Q‐PCR Master Mix (Gendepot, USA) and the following primers: IL‐4 (forward; f): 5'‐AACAGCCTCACAGAGCAGAAGAC‐3', IL‐4 (reverse; r): 5'‐GTGTTCTTGGAGGCAGCAAAG‐3'; IL‐5 (f): 5'‐TCTACTCATCGAACTCTGCTGA‐3', IL‐5 (r): 5'‐CCCTTGCACAGTTTGACTCTC‐3'; IL‐25 (f): 5'‐TTGTTTGTTTACTCATCACTCAG‐3', IL‐25 (r): 5'‐TCCTCCTCAGAATCATCCA‐3'; and IL‐33 (f): 5'‐TGTCACATTGGGCAAAGTT‐3', IL‐33 (r): 5'‐CAGTAAGCAGTGTTATCAGGAA‐3'. The mRNA expression was normalized using GAPDH (f): 5'‐ACCCACTCCTCCACCTTTG‐3' and GAPDH (r): 5'-ACCCACTCCTCC ACCTTTG‐3'. Relative quantitative expression was analyzed using the 2‐ΔΔCT method and determined as a fold change of the TNF-α/IFN-γ-treated control [19].
Protein Extraction and Western Blotting
Protein lysates were obtained using radioimmunoprecipitation assay (RIPA) cell lysis buffer (Gendepot) containing phosphatase and protease inhibitors (Gendepot). The extracted proteins were quantified at 1 μg/μl using Bradford (Coomassie) reagent (Gendepot). The protein samples were prepared in a 5X sample buffer (iNtron) and heated at 85°C for 10 min. Western blotting was performed using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS‐PAGE) [20]. The samples were loaded onto 10% Tris–glycine gels and electrophoresed in Tris/Glycine/SDS buffer (Bio-Rad). The separated proteins were transferred to polyvinylidene difluoride (PVDF) membranes (Millipore, USA) and washed with TBS-Tween buffer (TBS-T, Gendepot). After blocking with the Smart‐Block 5 min‐Fast Blocking Buffer (Biomax, Republic of Korea), the membranes were incubated with primary antibodies, phosphorylated (p)‐IκB, IκB, p‐nuclear factor kappa B (NF‐κB) p65, extracellular signal‐regulated kinases (ERK), p‐ERK, p‐c‐Jun N‐terminal kinase (JNK), p38, and p‐p38 (Cell Signaling Technology, USA), and NF-κB p65, JNK, and β‐actin (USA), diluted to 1:1000 overnight at 4°C. After washing with TBS‐T, membranes were incubated with HRP‐conjugated secondary antibodies (1:5000, Gendepot) for 1 h. The western blot images were developed using the LuminoGraph III Lite (Atto Corp., Japan) with the EzWest Lumi Plus (Atto), and density graph analysis was performed using the CS Analyzer 4 (Atto).
Gas Chromatograpy/Mass Spectrometry (GC/MS) Analysis of SCFAs
SCFAs in SL were analyzed using GC–MS (QP2020 NXW/ORP230, Shimadzu, Japan) equipped with a Stabilwax-DA column (60 m × 0.32 mm × 0.25 μm, Shimadzu) according to a previous study [21]. GC analysis was conducted in splitless mode using helium (flow rate, 2 ml/min) as the carrier gas. The oven temperature was held at 50°C for 2 min and then ramped up to 200°C at 10°C/min. This temperature was held for 5 min. The ionization was operated using the following settings: electron energy = 70 eV, ion source heater temperature = 200°C and 150°C, and scan speed = 0.2 s/scan. The analytes were monitored in the selected ion monitor (SIM) mode. Acetic acid, propionic acid, and butyric acid (Sigma-Aldrich) were used as standards and the quantitative analysis in CFS was calculated using the standard curve.
Profiling of Carbohydrate Fermentation and Enzymatic Activity
Carbohydrate fermentation of LAB was measured using the API 50 CHL Kit (BioMerieux, France) [22]. Briefly, LAB strains were grown on MRS agar (BD Biosciences) plates for 18 h, and the colonies were adjusted in API suspension medium (BioMerieux) to 2.0 McFarland. The suspended LAB strains were inoculated into strips and incubated at 37°C for 48 h. Meanwhile, the enzymatic activities of LAB were confirmed using the API ZYM Enzyme System (BioMerieux) [23]. After streaking on MRS medium, a colony was cultured at 37°C for 18 h, and the turbidity was adjusted to 5.0–6.0 McFarland in API suspension medium (BioMerieux). The suspension medium was dispensed on each enzyme of the API ZYM strip and incubated at 37°C for 4 h. The results were confirmed by the color changes.
Cytotoxicity to HT‐29 Cells
Cytotoxicity was estimated using the MTT assay [17]. HT‐29 cells were seeded at a density of 2.5 × 104 cells/well in 96‐well plates. LAB were treated at 106, 107, and 108 CFU/ml for 24 h, and then the culture medium was removed and MTT solution (0.25 mg/ml) was added to each cell for 2 h. The formazan crystals, formed by MTT solution, were dissolved in DMSO (150 μl). Absorbance at 550 nm was measured using a microplate spectrophotometer (Biotek).
Hemolytic and Bile Salt Hydrolase (BSH) Activity
Hemolytic activity was determined using Columbia agar (Oxoid, UK) containing 5% sheep blood (MBcell, Korea) [24]. LAB strains were streaked and cultured at 37°C for 48 h. Hemolysis was observed as halo zones around the colony: (1) alpha hemolysis appeared green in the medium, suggesting partial hemolysis; (2) beta hemolysis showed a clear zone, indicating destroyed red blood cells; and (3) gamma hemolysis did not show any reaction in the surrounding medium, indicating a lack of hemolysis [25]. Meanwhile, BSH enzyme activity of the LAB strains was determined according to a previous report [26]. Briefly, each strain was cultured on MRS agar containing 0.5% taurodeoxycholic acid hydrate (Sigma-Aldrich) and incubated at 37°C under anaerobic conditions for 48 h. BSH activity was assessed with the appearance of a precipitation zone surrounding the colony.
Survival in Gastrointesinal Tract (GIT) Conditions
Survival under conditions similar to those in the GIT environment was assessed as previously described [27]. The viability of LAB was measured after 2 h in PBS (pH 2.5) with 0.3% pepsin (Sigma-Aldrich) as gastric fluids at 37°C, and after 2.5 h in PBS (pH 7.4) with 1% pancreatin‐bile salt (Sigma-Aldrich) as intestinal fluids at 37°C. The live colony count was performed using an MRS agar plate.
Adhesion to HT‐29 Cells
The adhesion test to HT‐29 cells, a human colon adenocarcinoma cell line, was performed as reported previously with some modifications [28]. Briefly, HT‐29 cells were sub‐cultured at 1.5 × 105 cells/well in 12‐well plates and grown at 37°C in a humidified atmosphere of 5% CO2 until they formed a monolayer. The cells were then treated with LAB (1 × 108 CFU/ml) for 4 h. After treatment, the HT‐29 cells were washed three times with PBS and then lysed with 1 ml of PBS. The cell lysate was diluted with buffered peptone water (Oxoid), plated on MRS agar, and incubated at 37°C for 48 h. Adhesion ability (%) was calculated using the following equation: log (adherent counts of LAB) CFU/ml/log (initial counts of LAB) CFU/ml × 100.
Statistical Analysis
Results are expressed as mean ± standard error of the mean (SEM). Statistical significance was analyzed using one-way analysis of variance (ANOVA) followed by post hoc verification with Duncan’s multiple comparison tests (SPSS Statistics ver. 21, IBM, USA).
Results
SL Suppressed Chemokines in TNF-α/ IFN-γ‐Induced HaCaT Keratinocytes
HaCaT keratinocytes were treated with various concentrations of SL to optimize the concentration. The results showed no toxicity at 5% SL (Fig. 1A). As shown in Fig. 1B, 5% SL was not cytotoxic to TNF-α/IFN-γ‐induced HaCaT keratinocytes; therefore, further experiments were conducted at this concentration. TNF-α/IFN-γ-treated HaCaT keratinocytes showed significantly upregulated TARC levels (Fig. 1C). Conversely, treatment with SL from LAB and TNF-α/IFN-γ markedly decreased TARC levels (
-
Figure 1. Effect of SL (5%) from LAB on cell viability (A), expression of TARC (B) and MDC (C) in TNF-α/IFN- γ‐induced HaCaT keratinocytes.
The bars indicate the mean ± SEM. Different letters (a–f) show significant difference at
p < 0.05 (n = 3). *T+I, TNF-α/IFN-γ only treated group.
Inhibition of Cytokines Related to Immune Response by SL in TNF-α/IFN-γ‐Induced HaCaT Keratinocytes
To determine the effect of different SL on cytokines, IL‐4, IL‐5 , IL‐25, IL‐33, and thymic stromal lymphopoietin (TSLP) were analyzed using real-time PCR and ELISA. As shown in Fig. 2A, IL‐4 and IL‐5 mRNA levels were significantly elevated in TNF-α/IFNγ-induced HaCaT keratinocytes (
-
Figure 2. Effect of SL (5%) from LAB strains on
IL‐4 ,IL‐5 mRNA expression (A), TSLP (B) and IL‐25, IL‐33 mRNA expression (C) in TNF-α/IFN-γ‐induced HaCaT keratinocytes. The bars indicate the mean ± SEM. Different letters (a–e) show significant difference atp < 0.05 (n = 3). *T+I, TNF-α/IFN-γ only treated group.
Effect of SL on NF‐κB/MAPK Signaling Pathways in TNF-α/ IFN-γ‐Induced HaCaT Keratinocytes
To investigate the effect of SL on the NF-κB signaling pathway, p‐IκB and p‐NF‐κB were analyzed using western blotting (Fig. 3). Increased p‐IκB and p‐NF‐κB levels were observed in TNF-α/IFN-γ-induced HaCaT keratinocytes (
-
Figure 3. Effect of SL (5%) from LAB strains on the phosphorylation of nuclear factor kappa B (Nf‐κB) signaling pathway in TNF-α/IFN-γ‐induced HaCaT keratinocytes.
The protein expression of IκB (A) and Nf‐κB (B) were normalized to non‐phosphorylated protein. The bars indicate the mean ± SEM. Different letters (a–d) show significant difference at
p < 0.05 (n = 3). *T+I, TNF-α/IFN-γ only treated group.
-
Figure 4. Modulation of the mitogen‐activated protein kinase (MAPK) signaling by SL (5%) from LAB strains in TNF-α/IFN-γ‐induced HaCaT keratinocytes.
Representative western blot images of p‐ERK/ERK (A), p‐JNK/JNK (B), and p‐p38/p38 (C) expression was shown. The protein expression was normalized to non‐phosphorylated protein. The bars indicate the mean ± SEM. Different letters (a–d) show significant difference at
p < 0.05 (n = 3). *T+I, TNF-α/IFN-γ only treated group.
Analysis of SCFAs in SL from L. rhamnosus MG4644, L. paracasei MG4693, and Lc. lactis MG5474
To analyze SCFAs, which are major metaboiltes of probiotics produced by each strain, the contents of acetic, propionic and butyric acid in SL were measured using GC-MS (Table 2). The calibration curve formulas of acetic, propionic and butyric acid were confirmed as y = 35590x + 44682, y = 105472x + 10648, and y = 241306x + 3248.1, respectively, and through this, quantitative analysis of SCFA in SL was performed. All three strains were confirmed to have high levels of acetic acid content; however,
-
Table 2 . SCFA contents in CFS of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474..LAB SCFAs (mg/l) Acetic acid Propionic acid Butyric acid Total MG4644 2987.00 ± 65.79ab 1.49 ± 0.06a 5.37 ± 0.04a 2993.85 ± 65.69ab MG4693 3112.43 ± 79.35a 1.29 ± 0.11a 4.83 ± 0.22a 3118.55 ± 79.68a MG5474 2643.22 ± 136.40b 1.44 ± 0.03a 4.97 ± 0.19a 2649.63 ± 136.63b Data are presented as mean ± SEM. Different letters (a–d) show significant difference at
p < 0.05 (n = 3)..
Probiotic Properties of L. rhamnosus MG4644, L. paracasei MG4693, and Lc. lactis MG5474
As shown in the carbohydrate fermentation profiles, all three strains fermented various sugars (Table 3). We found that all strains utilized L-arabinose, methyl-β-D-xyloside, D-galactose, D-glucose, D-fructose, methyl-α-D-glucoside, arbutin, esculin, salicin, D-cellobiose, D-maltose, D-melibiose, and D-sucrose, and did not utilize glycerol, D-xylose, L-xylose, D-adonitol, D-sorbitol, starch, glycogen, D-turanose, D-tagatose, L-fucose, D-arabitol, gluconate, and 2-keto-gluconate. Each strain differed in its ability to utilize the remaining carbohydrates. Based on these results, the three strains were identified as
-
Table 3 . Carbohydrate fermentation profiles of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474..No. Type of testa MG4644 MG4693 MG5474 No. Type of test MG4644 MG4693 MG5474 1 Control - - - 26 Esculin + + + 2 Glycerol - - - 27 Salicin + + + 3 Erythritol - - - 28 D-Cellobiose + + + 4 D-Arabinose + - - 29 D-Maltose + + + 5 L-Arabinose - - + 30 D-Lactose + + + 6 D-Ribose + + + 31 D-Melibiose - - + 7 D-Xylose - - + 32 D-Sucrose + + + 8 L-Xylose - - - 33 D-Trehalose + + + 9 D-Adonitol - - - 34 Inulin - + - 10 Methyl-β-D-xyloside - - - 35 D-Melezitose + + - 11 D-Galactose + + + 36 D-Raffinose - - + 12 D-Glucose + + + 37 Starch - - + 13 D-Fructose + + + 38 Glycogen - - - 14 D-Mannose + + + 39 Xylitol - - - 15 L-Sorbose + - - 40 Gentiobiose + - + 16 L-Rhamnose + - - 41 D-Turanose + + - 17 Dulcitol + - - 42 D-Lyxose - - - 18 Inositol + - - 43 D-Tagatose + + - 19 D-Mannitol + + - 44 D-Fucose - - - 20 D-Sorbitol + - - 45 L-Fucose + - - 21 Methyl-α-D-mannoside - - - 46 D-Arabitol - - - 22 Methyl-α-D-glucoside + + - 47 L-Arabitol - - - 23 N-Acetylglucosamine + + + 48 Gluconate + + + 24 Amygdalin + - - 49 2-Keto-gluconate - - - 25 Arbutin + - + 50 5-Keto-gluconate - - - aThe mark of the result tests: -, not growth; +, growth..
-
Table 4 . Enzymatic profile of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474..Enzyme assayed fora MG4644 MG4693 MG5474 Control (Negative) 0 0 0 Alkaline phosphatase 1 0 0 Esterase (C4) 3 3 0 Esterase Lipase (C8) 2 3 0 Lipase (C14) 0 0 0 Leucine arylamidase 5 5 5 Valine arylamidase 5 5 2 Crystine arylamidase 2 1 2 Trypsin 0 0 0 α-Chymotrypsin 1 0 1 Acid phosphatase 2 3 3 Naphtol-AS-BI-phosphohydrolase 5 4 2 α-Galactosidase 0 0 0 β-Galactosidase 3 4 0 β-Glucuronidase 0 0 0 α-Glucosidase 2 4 0 β-Glucosidase 5 0 0 N–acetyl -β- glucosaminidase 0 1 0 α-Mannosidase 0 0 0 α-Fucosidase 2 0 0 aData are indicated on scale of 0 (no reaction) to 5 (maximum activity)..
Safety of L. rhamnosus MG4644, L. paracasei MG4693, and Lc. lactis MG5474
To confirm the safety of
-
Figure 5. Hemolysis (A), BSH activity (B), and cytotoxicity of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474 in HT‐29 cells (C). HT‐29 cells were treated withL. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474 (106, 107, and 108 cells/ml). Data are presented as the mean±SEM (n =3), and have no statistical significance.
Stability in Simulated GIT and Adhesion to HT‐29 Cells of L. rhamnosus MG4644, L. paracasei MG4693, and Lc. lactis MG5474
The stability in simulated GIT of
-
Figure 6. Cytotoxicity in HT‐29 cells (A), hemolysis (B), and BSH activity (C) of
L. rhamnosus MG4644,L. paracasei MG4693, and Lc. lactis MG5474. HT‐29 cells were treated withL. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474 (106, 107, and 108 cells/ml). Data are presented as the mean ± SEM (n = 3), and have no statistical significance.
Discussion
The gut-skin axis is currently being investigated based on studies showing that the immune regulation ability of the gut microbiome affects this distant organ [30]. Studies have reported that the skin and intestines have many similarities, and that skin conditions can be affected by changes in gut microbes [30]. The intestinal microflora acts on AD in two directions: (1) directly by producing metabolites such as SCFAs and delivering them to the skin through the blood vessels, and/or (2) indirectly by controlling the microbiota, which helps balance the immune system [31]. In a clinical study,
The stimulation of TNF-α and IFN-γ results in the expression of many cytokines (IL family and TSLP) and chemokines (MDC and TARC) in keratinocytes [36]. TARC and MDC, which are specific ligands for the CC motif chemokine ligand 4 (CCR4) expressed by Th2 cells, have been reported to correlate with the pathogenesis of AD. In our study, most SL downregulated MDC and TARC levels, which were elevated by TNF-α/IFN-γ in keratinocytes. The incidence of AD-like inflammation can be improved by regulating cytokines involved in T-helper cell type (Th)-2 and Th-1/Th-2 ratio [15]. Inhibition of Th2 cytokines such as IL-4 and IL-5 is known to block the progression of AD-like inflammation [11]. TSLP, which induces Th2 differentiation and produces Th2 cytokines, contributes to pruritus in keratinocytes of patients with AD [10]. In the present study, the mRNA expression of IL-4, IL-5, and TSLP were most inhibited by
Activation of NF-κB and MAPKs signaling pathways related to inflammation in keratinocytes is induced by cross-linking with TNF-α and IFN-γ [9]. NF-κB, stimulated by the degradation of IκB, is a major transcription factor involved in the inflammatory responses in AD [35]. Phosphorylated NF-κB translocates to the nucleus and produces cytokines and chemokines to regulate inflammatory and immune responses [36]. In our results, SL significantly diminished p‐IκB and p‐NF‐κB increased by TNF-α/IFN-γ in keratinocytes. Furthermore, NF-κB is regulated by the MAPK signaling pathway [36]. A previous study reported that inhibition of p‐p38 and p‐ERK attenuates skin inflammation [9]. This study showed that SL significantly inhibited p‐p38 and p‐ERK in TNF-α/IFN-γ-induced HaCaT keratinocytes. In summary,
The FAO/WHO Working Group reported that LAB strains should demonstrate probiotic properties, including tolerance under gastrointestinal tract (GIT) conditions and bile salt hydrolase (BSH) de‐conjugation, adhesion to intestinal epithelial cells, and safety (hemolysis and cytotoxicity) [7]. Safety evaluation of LAB strains is the first priority for their use as probiotics because they are rarely used as causative agents of potential opportunistic infections [41]. The MTT assay can measure mitochondrial activity in living cells and is therefore used to measure the cytotoxic effects of samples in vitro [42]. In addition, this assay is widely used as a cytotoxic screening method to evaluate the safety of probiotics [43]. Our results showed that all three strains were non-toxic to intestinal epithelial cells. In addition, hemolysis and BSH activities are considered important safety traits for probiotics [41]. As bile salts are synthesized from cholesterol, some LAB with BSH activity have been suggested to prevent hypercholesterolemia [44]. However, excessive BSH activity can have detrimental effects on the host by causing lipid dyspepsia and impairing colonic mucosal function, potentially leading to the formation of gallstones [45]. In this study,
For consumption as a probiotic, stability in the GIT in the host is important [13]. In other words, probiotics must tolerate bile salt concentrations, enzymes such as pepsin and pancreatin, and low pH (2.5–3.5) [46]. This study found that
Conclusion
In the current study,
Conflict of Interest
The authors have no financial conflicts of interest to declare.
Fig 1.
Fig 2.
Fig 3.
Fig 4.
Fig 5.
Fig 6.
-
Table 1 . The isolates and accession numbers of LAB strains used in this study..
LAB Accession No. Human L. rhamnosus MG4288MW947157.1 L. rhamnosus MG4643ON668169.1 L. rhamnosus MG4644ON668170.1 L. paracasei MG4693OP077096.1 Food Lc. lactis MG5474ON619520.1 Lc. lactis MG5604ON705160.1 L. reuteri MG5462ON619508.1
-
Table 2 . SCFA contents in CFS of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474..LAB SCFAs (mg/l) Acetic acid Propionic acid Butyric acid Total MG4644 2987.00 ± 65.79ab 1.49 ± 0.06a 5.37 ± 0.04a 2993.85 ± 65.69ab MG4693 3112.43 ± 79.35a 1.29 ± 0.11a 4.83 ± 0.22a 3118.55 ± 79.68a MG5474 2643.22 ± 136.40b 1.44 ± 0.03a 4.97 ± 0.19a 2649.63 ± 136.63b Data are presented as mean ± SEM. Different letters (a–d) show significant difference at
p < 0.05 (n = 3)..
-
Table 3 . Carbohydrate fermentation profiles of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474..No. Type of testa MG4644 MG4693 MG5474 No. Type of test MG4644 MG4693 MG5474 1 Control - - - 26 Esculin + + + 2 Glycerol - - - 27 Salicin + + + 3 Erythritol - - - 28 D-Cellobiose + + + 4 D-Arabinose + - - 29 D-Maltose + + + 5 L-Arabinose - - + 30 D-Lactose + + + 6 D-Ribose + + + 31 D-Melibiose - - + 7 D-Xylose - - + 32 D-Sucrose + + + 8 L-Xylose - - - 33 D-Trehalose + + + 9 D-Adonitol - - - 34 Inulin - + - 10 Methyl-β-D-xyloside - - - 35 D-Melezitose + + - 11 D-Galactose + + + 36 D-Raffinose - - + 12 D-Glucose + + + 37 Starch - - + 13 D-Fructose + + + 38 Glycogen - - - 14 D-Mannose + + + 39 Xylitol - - - 15 L-Sorbose + - - 40 Gentiobiose + - + 16 L-Rhamnose + - - 41 D-Turanose + + - 17 Dulcitol + - - 42 D-Lyxose - - - 18 Inositol + - - 43 D-Tagatose + + - 19 D-Mannitol + + - 44 D-Fucose - - - 20 D-Sorbitol + - - 45 L-Fucose + - - 21 Methyl-α-D-mannoside - - - 46 D-Arabitol - - - 22 Methyl-α-D-glucoside + + - 47 L-Arabitol - - - 23 N-Acetylglucosamine + + + 48 Gluconate + + + 24 Amygdalin + - - 49 2-Keto-gluconate - - - 25 Arbutin + - + 50 5-Keto-gluconate - - - aThe mark of the result tests: -, not growth; +, growth..
-
Table 4 . Enzymatic profile of
L. rhamnosus MG4644,L. paracasei MG4693, andLc. lactis MG5474..Enzyme assayed fora MG4644 MG4693 MG5474 Control (Negative) 0 0 0 Alkaline phosphatase 1 0 0 Esterase (C4) 3 3 0 Esterase Lipase (C8) 2 3 0 Lipase (C14) 0 0 0 Leucine arylamidase 5 5 5 Valine arylamidase 5 5 2 Crystine arylamidase 2 1 2 Trypsin 0 0 0 α-Chymotrypsin 1 0 1 Acid phosphatase 2 3 3 Naphtol-AS-BI-phosphohydrolase 5 4 2 α-Galactosidase 0 0 0 β-Galactosidase 3 4 0 β-Glucuronidase 0 0 0 α-Glucosidase 2 4 0 β-Glucosidase 5 0 0 N–acetyl -β- glucosaminidase 0 1 0 α-Mannosidase 0 0 0 α-Fucosidase 2 0 0 aData are indicated on scale of 0 (no reaction) to 5 (maximum activity)..
References
- Torres T, Ferreira E, Gonçalo M, Mendes-Bastos P, Selores M, Filipe P. 2019. Update on atopic dermatitis.
Acta Medica Port. 32 : 606-613. - Yoo J, Koo HYR, Han K, Lee YB. 2022. Impairment of quality of life and mental health status in adult-onset atopic dermatitis.
Ann. Dermatol. 34 : 278-286. - Ha J, Lee SW, Yon DK. 2020. Ten-year trends and prevalence of asthma, allergic rhinitis, and atopic dermatitis among the Korean population, 2008-2017.
Clin. Exp. Pediatr. 63 : 278-283. - Manjelievskaia J, Boytsov N, Brouillette MA, Onyekwere U, Pierce E, Goldblum O,
et al . 2021. The direct and indirect costs of adult atopic dermatitis.J. Manag. Care Spec. Pharm. 27 : 1416-1425. - Albader SS, Alharbi AA, Alenezi RF, Alsaif FM. 2019. Dupilumab side effect in a patient with atopic dermatitis: a case report study.
Biologics 13 : 79-82. - Lee JY, Kim Y, Kim J-I, Lee H-Y, Moon G-S, Kang C-H. 2022. Improvements in human keratinocytes and antimicrobial effect mediated by cell-free supernatants derived from probiotics.
Fermentation 8 : 332. - Bitschar K, Wolz C, Krismer B, Peschel A, Schittek B. 2017. Keratinocytes as sensors and central players in the immune defense against
Staphylococcus aureus in the skin.J. Dermatol. Sci. 87 : 215-220. - Albanesi C, Scarponi C, Giustizieri ML, Girolomoni G. 2005. Keratinocytes in inflammatory skin diseases.
Curr. Drug Targets Inflamm. Allergy 4 : 329-334. - Cho SH, Kim HS, Lee W, Han EJ, Kim SY, Fernando IS,
et al . 2020. Eckol fromEcklonia cava ameliorates TNF-α/IFN-γ-induced inflammatory responses via regulating MAPKs and NF-κB signaling pathway in HaCaT cells.Int. Immunopharmacol. 82 : 106146. - Chieosilapatham P, Kiatsurayanon C, Umehara Y, Trujillo-Paez J, Peng G, Yue H,
et al . 2021. Keratinocytes: Innate immune cells in atopic dermatitis.Clin. Expe. Immunol. 204 : 296-309. - Yan F, Li F, Liu J, Ye S, Zhang Y, Jia J,
et al . 2020. The formulae and biologically active ingredients of Chinese herbal medicines for the treatment of atopic dermatitis.Biomed. Pharmacother. 127 : 110142. - Edalati E, Saneei B, Alizadeh M, Hosseini SS, Bialvaei AZ, Taheri K. 2019. Isolation of probiotic bacteria from raw camel's milk and their antagonistic effects on two bacteria causing food poisoning.
New Microb. New Infect. 27 : 64-68. - Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B,
et al . 2014. The international scientific association for robiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic.Nat. Rev. Gastroenterol. Hepatol. 11 : 506-514. - Lizardo MV, Tavaria FK. 2022. Probiotics and skin health, pp. 389-405. Probiotics, Ed. Elsevier.
- Rusu E, Enache G, Cursaru R, Alexescu A, Radu R, Onila O,
et al . 2019. Prebiotics and probiotics in atopic dermatitis.Exp. Ther. Med. 18 : 926-931. - Lew LC, Liong MT. 2013. Bioactives from probiotics for dermal health: functions and benefits.
J. Appl. Microbiol. 114 : 1241-1253. - Lee JY, Kang JH, Jung YR, Kang CH. 2022.
Lactobacillus gasseri MG4247 andLacticaseibacillus paracasei MG4272 and MG4577 modulate allergic inflammatory response in RAW 264.7 and RBL-2H3 cells.Probiotics Antimicrob. Proteins 31 : 1-10. - Tolosa L, Donato MT, Gómez-Lechón MJ. 2015. General cytotoxicity assessment by means of the MTT assay, pp. 333-348. Protocols in in vitro hepatocyte research, Ed. Springer.
- Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method.
Methods 25 : 402-408. - Bass JJ, Wilkinson DJ, Rankin D, Phillips BE, Szewczyk NJ, Smith K,
et al . 2017. An overview of technical considerations for Western blotting applications to physiological research.Scand. J. Med. Sci. Sports 27 : 4-25. - Park JY, Lee JY, Kim Y, Kang CH. 2023.
Latilactobacillus sakei Wikim0066 protects skin through MMP regulation on UVB-irradiated in vitro and in vivo model.Nutrients 15 : 726. - Syed Yaacob SN, Huyop F, Kamarulzaman Raja Ibrahim R, Wahab RA. 2018. Identification of
Lactobacillus spp. andFructobacillus spp. isolated from fresh Heterotrigona itama honey and their antagonistic activities against clinical pathogenic bacteria.J. Apic. Res. 57 : 395-405. - Kang CH, Kim Y, Han SH, Jeong Y, Park H, Paek NS. 2019. Probiotic properties of bifidobacteria isolated from feces of infants.
J. Milk Sci. Biotechnol. 37 : 40-48. - Yasmin I, Saeed M, Khan WA, Khaliq A, Chughtai MFJ, Iqbal R,
et al . 2020. In vitro probiotic potential and safety evaluation (hemolytic, cytotoxic activity) ofBifidobacterium strains isolated from raw camel milk.Microorganisms 8 : 354. - Buxton R. 2005. Blood agar plates and hemolysis protocols. American Society for Microbiology. pp. 1-9.
- Shehata M, El Sohaimy S, El-Sahn MA, Youssef M. 2016. Screening of isolated potential probiotic lactic acid bacteria for cholesterol lowering property and bile salt hydrolase activity.
Ann. Agric. Sci. 61 : 65-75. - Kim S, Lee JY, Jeong Y, Kang CH. 2022. Antioxidant activity and probiotic properties of lactic acid bacteria.
Fermentation 8 : 29. - Fonseca HC, de Sousa Melo D, Ramos CL, Dias DR, Schwan RF. 2021. Probiotic properties of lactobacilli and their ability to inhibit the adhesion of enteropathogenic bacteria to Caco-2 and HT-29 cells.
Probiotics Antimicrob. Proteins 13 : 102-112. - Cheng KW, Tseng CH, Chen IJ, Huang BC, Liu HJ, Ho KW,
et al . 2022. Inhibition of gut microbial β-glucuronidase effectively prevents carcinogen-induced microbial dysbiosis and intestinal tumorigenesis.Pharmacol. Res. 177 : 106115. - De Pessemier B, Grine L, Debaere M, Maes A, Paetzold B, Callewaert C. 2021. Gut-skin axis: current knowledge of the interrelationship between microbial dysbiosis and skin conditions.
Microorganisms 9 : 353. - Lee SY, Lee E, Park YM, Hong SJ. 2018. Microbiome in the gut-skin axis in atopic dermatitis.
Allergy, Asthma Immunol. Res. 10 : 354-362. - Lolou V, Panayiotidis MI. 2019. Functional role of probiotics and prebiotics on skin health and disease.
Fermentation 5 : 41. - De Vuyst E, Salmon M, Evrard C, Lambert de Rouvroit C, Poumay Y. 2017. Atopic dermatitis studies through in vitro models.
Front. Med. 4 : 119. - Song HK, Park SH, Kim HJ, Jang S, Kim T. 2022.
Spatholobus suberectus dunn water extract ameliorates atopic dermatitis-like symptoms by suppressing proinflammatory chemokine production in vivo and in vitro.Front. Pharmacol. 13 : 919230. - Hwang DH, Koh PO, Kang C, Kim E. 2021. Rosa davurica Pall. improves DNCB-induced atopic dermatitis in mice and regulated TNF-alpha/IFN-gamma-induced skin inflammatory responses in HaCaT cells.
Phytomedicine 91 : 153708. - Yang CC, Hung YL, Ko WC, Tsai YJ, Chang JF, Liang CW,
et al . 2021. Effect of neferine on DNCB-induced atopic dermatitis in HaCaT cells and BALB/c mice.Int. J. Mol. Sci. 22 : 8237. - Sebastian K, Borowski A, Kuepper M, Friedrich K. 2008. Signal transduction around thymic stromal lymphopoietin (TSLP) in atopic asthma.
Cell Commun. Signal. 6 : 5. - Divekar R, Kita H. 2015. Recent advances in epithelium-derived cytokines (IL-33, IL-25 and TSLP) and allergic inflammation.
Curr. Opin. Allergy Clin. Immunol. 15 : 98. - Xiao X, Hu X, Yao J, Cao W, Zou Z, Wang L,
et al . 2022. The role of short-chain fatty acids in inflammatory skin diseases.Front. Microbiol. 13 : 1083432. - Kang CH, Kim JS, Park HM, Kim S, Paek NS. 2021. Antioxidant activity and short-chain fatty acid production of lactic acid bacteria isolated from Korean individuals and fermented foods.
3 Biotech. 11 : 217. - Aryantini NPD, Yamasaki E, Kurazono H, Sujaya IN, Urashima T, Fukuda K. 2017. In vitro safety assessments and antimicrobial activities of
Lactobacillus rhamnosus strains isolated from a fermented marés milk.Anim. Sci. J. 88 : 517-525. - Meerloo JV, Kaspers GJ, Cloos J. 2011. Cell sensitivity assays: the MTT assay, pp. 237-245. Cancer cell culture, Ed. Springer.
- Ranjan KG, Sankar GG, Raju D. 2020. In vitro evaluation of antibacterial, cytotoxic and adherence studies of selected commercial probiotics.
J. Pure Appl. Microbiol. 14 : 2085-2091. - Khare A, Gaur S. 2020. Cholesterol-lowering effects of
Lactobacillus species.Curr. Microbiol. 77 : 638-644. - Byakika S, Mukisa IM, Byaruhanga YB, Muyanja C. 2019. A review of criteria and methods for evaluating the probiotic potential of microorganisms.
Food Rev. Int. 35 : 427-466. - Casarotti SN, Todorov SD, Penna ALB. 2015. Effect of different matrices on probiotic resistance to in vitro simulated gastrointestinal conditions.
Int. J. Dairy Technol. 68 : 595-601. - Jovanović JN, Nikolić B, Šeatović S, Zavišić G, Mitić-Ćulafić D, Vuković-Gačić B,
et al . 2015. Characterization of some potentially probioticLactobacillus strains of human origin.Food Sci. Biotechnol. 24 : 1781-1788. - Verhoeckx K, Cotter P, López-Expósito I, Kleiveland C, Lea T, Mackie A,
et al . 2015. The impact of food bioactives on health: in vitro and ex vivo models.Cham CH : Springer.